US20070244067A1 - Devices and processes for distribution of genetic material to mammalian limb - Google Patents

Devices and processes for distribution of genetic material to mammalian limb Download PDF

Info

Publication number
US20070244067A1
US20070244067A1 US11/694,140 US69414007A US2007244067A1 US 20070244067 A1 US20070244067 A1 US 20070244067A1 US 69414007 A US69414007 A US 69414007A US 2007244067 A1 US2007244067 A1 US 2007244067A1
Authority
US
United States
Prior art keywords
muscle
polynucleotide
dna
limb
expression
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US11/694,140
Other versions
US7642248B2 (en
Inventor
Vladimir Budker
Jon Wolff
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Arrowhead Madison Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/707,000 external-priority patent/US7507722B1/en
Priority claimed from US09/707,117 external-priority patent/US7396821B1/en
Priority claimed from US10/339,934 external-priority patent/US20070173465A9/en
Priority claimed from US10/429,478 external-priority patent/US7214369B2/en
Priority to US11/694,140 priority Critical patent/US7642248B2/en
Application filed by Individual filed Critical Individual
Publication of US20070244067A1 publication Critical patent/US20070244067A1/en
Assigned to MIRUS BIO CORPORATION reassignment MIRUS BIO CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WOLFF, JON A, BUDKER, TATYANA
Assigned to ROCHE MADISON INC. reassignment ROCHE MADISON INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: MIRUS BIO CORPORATION
Publication of US7642248B2 publication Critical patent/US7642248B2/en
Application granted granted Critical
Assigned to Arrowhead Madison Inc. reassignment Arrowhead Madison Inc. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: ROCHE MADISON INC.
Adjusted expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0075Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the delivery route, e.g. oral, subcutaneous

Definitions

  • the invention relates to devices and processes for use in biological systems. More particularly, processes that provide for the functional distribution of genetic material to mammalian cells are described.
  • Biotechnology includes the delivery of genetic information to a cell to express an exogenous nucleotide sequence, to inhibit, eliminate, augment, or alter expression of an endogenous nucleotide sequence, or to express a specific physiological characteristic not naturally associated with the cell.
  • Polynucleotides may be coded to express a whole or partial protein, or may be anti-sense, or non-viral DNA, or recombine with chromosomal DNA.
  • Delivery of a nucleic acid means to transfer a nucleic acid from a container outside a mammal to near or within the outer cell membrane of a cell in the mammal.
  • transfection is used herein, in general, as a substitute for the term delivery, or, more specifically, the transfer of a nucleic acid from directly outside a cell membrane to within the cell membrane.
  • the transferred (or transfected) nucleic acid may contain an expression cassette. If the nucleic acid is a primary RNA transcript that is processed into messenger RNA, a ribosome translates the messenger RNA to produce a protein within the cytoplasm.
  • nucleic acid If the nucleic acid is a DNA, it enters the nucleus where it is transcribed into a messenger RNA that is transported into the cytoplasm where it is translated into a protein. Therefore if a nucleic acid expresses its cognate protein, then it must have entered a cell. A protein may subsequently be degraded into peptides, which may be presented to the immune system.
  • the muscular dystrophies are a heterogeneous group of mostly inherited disorders characterized by progressive muscle wasting and weakness which eventually leads to death. In most in not all forms of MD, the disease is associated with either a non-functioning or malfunctioning protein due to the presence of a mutant or deleted gene (Hartigan-O'Connor 2000). Because of the nature of these diseases, few traditional treatments are available. However, because the genes and protein products that are responsible for most of the dystrophies have been identified, delivery of corrective genes offers a promising treatment.
  • striated muscle cells have a long life span, facilitating long term persistence of delivered genes.
  • DMD and MCDM it has been shown that gene replacement in striated muscle alone can alleviate the major features of the disease (Cox 1993, Kuang 1998).
  • Dytrophin positive fibers may possess a survival advantage over dystrophin negative fibers, suggesting that only a portion of the fibers need to receive the correcting polynucleotide (Morgan 1993).
  • Morgan 1993 the correcting polynucleotide
  • only 20% of the normal level of dystrophin is required to be asymptomatic. Thus, low level dystrophin expression in a majority of muscle fibers may be sufficient for elimination of symptoms (Phelps 1995).
  • a process for delivering a polynucleotide into a parenchymal cell of a mammal, comprising making a polynucleotide such as a nucleic acid, inserting the polynucleotide into a mammalian vessel, such as a blood vessel, increasing the permeability of the vessel, and delivering the polynucleotide to the parenchymal cell thereby altering endogenous properties of the cell.
  • Increasing the permeability of the vessel consists of increasing pressure against vessel walls and/or inhibiting the flow of fluid through the vessel.
  • an in vivo process for delivering a polynucleotide to a parenchymal cell of a mammal is described.
  • the polynucleotide is inserted into a blood vessel and interior blood flow is impeded by external application of compression and the polynucleotide is delivered to the parenchymal cell.
  • the polynucleotide may consist of naked DNA, a viral particle/vector, a non-viral vector or may be a blocking polynucleotide for preventing gene expression.
  • the parenchymal cell may consist of a muscle cell, such as a limb (leg or arm) muscle cell.
  • the process includes externally impeding interior blood flow by externally applying pressure to interior blood vessels such as compressing mammalian skin by applying a tourniquet over the skin.
  • Compressing mammalian skin also includes applying a cuff over the skin such as a sphygmomanometer.
  • an in vivo process for delivering a polynucleotide to a mammalian cell consists of inserting the polynucleotide into a blood vessel and applying pressure to one or more blood vessels. The pressure is applied externally to mammalian skin and the polynucleotide is delivered to the mammalian cell.
  • the process especially consists of a polynucleotide delivered to non-vascular (not of the smooth muscle cells surrounding a vessel) parenchymal cells.
  • a device for applying pressure to mammalian skin for in vivo delivery of a polynucleotide to a mammalian cell consists of a cuff, as defined in this specification, applied to mammalian skin to impede fluid flow through the vessel thereby increasing delivery efficiency of the polynucleotide to the mammalian cell.
  • Immunosuppression can be of long term or for a short duration. For short term duration, the immunosuppression is preferably supplied around the time of nucleic acid delivery. Immunosuppression can be accomplished by treatment with (combinations of) immunosuppressive drugs like cyclosporin A, ProGraf (FK506), corticosteroids, deoxyspergualin, and dexamethasone.
  • immunosuppressive drugs like cyclosporin A, ProGraf (FK506), corticosteroids, deoxyspergualin, and dexamethasone.
  • Other methods include blocking of immune cell activation pathways, for instance by treatment with (or expression of) an antibody directed against CTLA4; redirection of activated immune cells by treatment with (ore expression of) chemokines such as MIP-1a, MCP-1 and RANTES; and treatment with immunotoxins, such as a conjugate between anti-CD3 antibody and diphtheria toxin.
  • chemokines such as MIP-1a, MCP-1 and RANTES
  • immunotoxins such as a conjugate between anti-CD3 antibody and diphtheria toxin.
  • the process may be used to deliver a therapeutic polynucleotide to a muscle cell for the treatment of vascular disease or occlusion.
  • the delivered polynucleotide can express a protein or peptide that stimulates angiogenesis, vasculogenesis, arteriogenesis, or anastomoses to improve blood flow to a tissue.
  • the gene may be selected from the list comprising: VEGF, VEGF II, VEGF-B, VEGF-C, VEGF-D, VEGF-E, VEGF 121 , VEGF 138 , VEGF 145 , VEGF 165 , VEGF 189 , VEGF 206 , hypoxia inducible factor 1 ⁇ (HIF 1 ⁇ ), endothelial NO synthase (eNOS), iNOS, VEFGR-1 (Flt1), VEGFR-2 (KDR/Flk1), VEGFR-3 (Flt4), neuropilin-1, ICAM-1, factors (chemokines and cytokines) that stimulate smooth muscle cell, monocyte, or leukocyte migration, anti-apoptotic peptides and proteins, fibroblast growth factors (FGF), FGF-1, FGF-1b, FGF-1c, FGF-2, FGF-2b, FGF-2c, FGF-3, FGF-3b, FGF-3c, F
  • the protein or peptide may be secreted or stay within the cell.
  • the gene may contain a sequence that codes for a signal peptide.
  • the delivered polynucleotide can also suppress or inhibit expression of an endogenous gene or gene product that inhibits angiogenesis, vasculogenesis, arteriogenesis or anastomosis formation. Multiple polynucleotides or polynucleotides containing more that one therapeutic gene may be delivered using the described process.
  • the gene or genes can be delivered to stimulate vessel development, stimulate collateral vessel development, promote peripheral vascular development, improve blood flow in a muscle tissue, or to improve abnormal cardiac function.
  • the gene or genes can also be delivered to treat peripheral circulatory disorders, myocardial disease, myocardial ischemia, limb ischemia, arterial occlusive disease, peripheral arterial occlusive disease, vascular insufficiency, vasculopathy, arteriosclerosis obliterans, thromboangiitis obliterans, atherosclerosis, aortitis syndrome, Behcet's disease, collagenosis, ischemia associated with diabetes, claudication, intermittent claudication, Raynaud disease, cardiomyopathy or cardiac hypertrophy.
  • the polynucleotide can be delivered to a muscle cell that is suffering from ischemia or a normal muscle cell.
  • the muscle cell can be a cardiac cell or a skeletal muscle cell.
  • a preferred skeletal muscle cell is a limb skeletal muscle cell.
  • the polynucleotides can also be delivered to cells in a tissue that is at risk of suffering from ischemia or a vascular disease or disorder.
  • the process may be used to deliver a therapeutic polynucleotide to a muscle cell for the treatment of muscular dystrophy.
  • the defective genes that cause MD are known for many forms of the disease. These defective genes either fail to produce a protein product, produce a protein product that fails to function properly, or produce a dysfunctional protein product that interferes with the proper function of the cell.
  • the polynucleotide may encode a therapeutically functional protein or a polynucleotide that inhibits production or activity of a dysfunctional protein.
  • the polynucleotide may be delivered to muscle cells of an MD patient for therapeutic treatment of the disease, for treatment of secondary manifestations of muscular dystrophy, or for other muscular atrophy or injury.
  • Genes that may be expressed from delivered polynucleotides, or inhibited by delivered polynucleotides, may be selected from the list comprising: dystrophin (Duchene's and Becker MD); dystrophin-associated glycoproteins ( ⁇ -sarcoglycan and ⁇ -sarcoglycan, limb-girdle MD 2E and 2F; ⁇ -sarcoglycan and ⁇ -sarcoglycan, limb-girdle MD 2D and 2C), utrophin, calpain (autosomal recessive limb-girdle MD type 2A), caveolin-3 (autosomal-dominant limb-girdle MD), laminin-alpha2 (merosin-deficient congenital MD), fukutin (Fukuyama type congenital MD) and emerin (Emery-Dreifuss MD), myotilin, lamin A/C, calpain-3, dysferlin, telethonin or
  • a polynucleotide expressing a protein beneficial to a patient suffering from muscular disease or injury or booster genes aimed at alleviating secondary defects of muscle disease may also be delivered to muscle cells of the patient.
  • Such genes may be selected from the list comprising: mini-agrin (to promote basement membrane formation), utrophin, laminin ⁇ 2, ⁇ 7 integrins, GalNac transferase, and ADAM12 (to promote cell adhesion and muscle stability), calpastatin (to protect against muscle necrosis), nitric oxide synthase (to ease inflammation), ADAM12 protein (meltrin alpha), IGF-1, dominant negative myostatin and myostatin inhibitors (to promote muscle regeneration and reduce fibrosis), TGF- ⁇ (to regulate muscle mass), Nitric Oxide Synthase (to reduce inflammation), actin, titin, muscle creatine kinase, troponin, growth factors (human growth factor and human growth hormone releasing hormone, and vascular endothelial growth factor (VEGF
  • Polynucleotides such as siRNAs and antisense oligonucleotides may also be delivered to create a myostatin blockade or to inhibit myostatin synthesis (to promote muscle growth), inhibit myogenin production (to increase muscle size) or to modify splicing of a defective endogenous gene.
  • the delivered polynucleotide may express a protein or peptide may be secreted or stay within the cell.
  • the gene may contain a sequence that codes for a signal peptide.
  • the delivered polynucleotide can also suppress or inhibit expression of an endogenous gene or gene product. Multiple polynucleotides or polynucleotides containing more than one therapeutic gene may be delivered using the described process.
  • FIG. 2A-2C Expression of ⁇ -galactosidase (light grey) and GFP (white) in rat muscle injected intraarterially at different times with the respective expression pDNAs.
  • Panel A 640 ⁇ magnification
  • Panels B and C are high power fields (1600 ⁇ magnification) that show an example of co-localization (B) and separate expression (C).
  • FIG. 3A-3C Muscle sections obtained 5 min (A and B) and 1 h (C) after 50 ⁇ g of Rh-pDNA in 10 ml of normal saline were injected within 7 sec into the femoral artery of rat without impeding the outflow (A) or impeding outflow (B and C). Arrows indicate Rh-pDNA between cells and arrowheads indicate pDNA inside myofibers. Magnification: 1260 ⁇ .
  • FIG. 4A-4C LacZ expression in mouse skeletal muscle seven days following intra-arterial injections of 100 ⁇ g pCI-LacZ (A) or pMI-DYS (B and C) in dystrophic mdx mouse (A and B) or normal ICR mouse (C).
  • FIG. 5 Intravascular injection of therapeutic genes into mammalian limbs. Immunohistochemical staining for human dystrophin expression in mdx4cv mouse gastrocnemius muscle (left panel) one week after intravenous injection of 300 ⁇ g of a pDNA human dystrophin expression vector in 0.6 ml of NSS (7.5 s injection). Staining in mdx4cv mice injected with pCI-Luc negative control vector is shown in the right panel.
  • FIG. 6A-6B Illustration of luciferase expression in leg muscles of dystrophic and normal dog after intra-arterial injection of pCI-Luc plasmid under elevated pressure.
  • Panel A shows expression distribution in normal dog.
  • Panel B shows expression distribution in dystrophic dog model.
  • FIG. 7 Paraffin cross sections of the Pronator quadratus muscles stained with hematoxylin and eosin and examined under light microscope.
  • Left panel Pronator quadratus muscle transfected with VEGF-165 plasmid.
  • Right panel Pronator quadratus muscle transfected with EPO plasmid.
  • Top left picture VEGF-165) demonstrates increased number of vessels and interstitial cells (presumably—endothelial cells), as compared to right picture (EPO-control), magnification ⁇ 200.
  • Bottom left picture (VEGF-165) demonstrates increased number of vessels, most small arteries and capillaries, as compare to right picture (EPO-control). Arrows indicate obvious vascular structures, magnification ⁇ 6300.
  • FIG. 8 Paraffin cross sections of the Pronator quadratus muscles immunostained for endothelial cell marker—CD31, and examined under confocal laser scanning microscope LSM 510, magnification ⁇ 400.
  • CD31 marker visualized with Cy3 (black), nuclei with nucleic acid stains To Pro-3.
  • Muscle fibers and red blood cells were visualized by 488 nm laser having autofluorescent emission.
  • Left picture Pronator quadratus muscle transfected with VEGF-165 plasmid, demonstrates increased of endothelial cells and small vessels, as compare to right picture (EPO-control). The number of CD31 positive cells was increased significantly in VEGF-165 transfected muscle by 61.7% (p ⁇ 0.001).
  • an intravascular route of administration allows a polynucleotide to be delivered to parenchymal cells in a more even distribution than direct parenchymal injections.
  • the efficiency of polynucleotide delivery and expression is increased by increasing the permeability of the tissue's blood vessel. Permeability is increased by one or more of the following: increasing the intravascular pressure, delivering the injection fluid rapidly (injecting the injection fluid rapidly), using a large injection volume, inhibiting vessel fluid flow, and increasing permeability of the vessel wall. Prior to insertion, subsequent to insertion, or concurrent with insertion the permeability of the vessel is increased using an exterior cuff thereby the genetic material is delivered to the parenchymal cell.
  • the described process may also be used repetitively in a single mammal. Multiple injections may be used to provide delivery to additional cells, tissues, or muscle groups, to increase delivery to a single tissue, or where multiple treatments are indicated. Multiple injections may be performed in different limbs of the same animal, within the same limb of the animal, within the same vein of the animal, within different veins in the animal (in the same or different limbs). The site of vessel occlusion may also be the same or different for multiple injections in the same animal.
  • cuff means a device for impeding fluid flow through mammalian internal blood vessels.
  • cuff refers specifically to a device applied exterior to the mammal's skin and touches the skin in a non-invasive manner.
  • the cuff is a device that applies external pressure to the mammalian skin and thereby pressure is applied internally to the blood vessel walls.
  • the vessel walls, in an area underneath the cuff, are forced to constrict in amount sufficient to impede fluid from flowing at a normal rate. Impeding fluid flow into and out of an area such as a limb, combined with injection of a solution containing polynucleotides, causes vascular pressure and vessel permeability to increase in the area.
  • a cuff which is normally used to measure blood pressure.
  • the sphygmomanometer is used to apply pressure to mammalian skin, around a limb, for the purpose of increasing vessel permeability when combined with solution injection into the vessel.
  • a tourniquet is another example.
  • a cuff or other external pressure device
  • a pharmaceutical or biologically-active agent such as papaverine
  • intravascular refers to an intravascular route of administration that enables a polymer, oligonucleotide, or polynucleotide to be delivered to cells more evenly distributed than direct injections.
  • Intravascular herein means within an internal tubular structure called a vessel that is connected to a tissue or organ within the body of an animal, including mammals.
  • a bodily fluid flows to or from the body part.
  • bodily fluid include blood, lymphatic fluid, or bile.
  • vessels include arteries, arterioles, capillaries, venules, sinusoids, veins, lymphatics, and bile ducts.
  • the intravascular route includes delivery through the blood vessels such as an artery or a vein.
  • Afferent blood vessels of organs are defined as vessels in which blood flows toward the organ or tissue under normal physiologic conditions.
  • Efferent blood vessels are defined as vessels in which blood flows away from the organ or tissue under normal physiologic conditions.
  • afferent vessels are known as coronary arteries, while efferent vessels are referred to as coronary veins.
  • Proximal is defined herein as a position nearer to the point of attachment or origin or central point, especially located nearer the center of the body, as the proximal part of a limb.
  • the proximal end of the femur is part of the hip joint
  • the knee is proximal to the toes
  • the shoulder is proximal to the elbow
  • the elbow is proximal to the hand.
  • the opposite of proximal is distal.
  • naked nucleic acid indicates that the nucleic acids are not associated with a transfection reagent or other delivery vehicle that is required for the nucleic acid to be delivered to a target cell.
  • a transfection reagent is a compound or compounds used in the prior art that mediates nucleic acid's entry into cells.
  • Expression cassette refers to a natural or recombinantly produced nucleic acid which is capable of expressing protein(s).
  • a DNA expression cassette typically includes a promoter (allowing transcription initiation), and a sequence encoding one or more proteins (transgene(s)).
  • the expression cassette may include transcriptional enhancers, locus control regions, matrix attachment regions, scaffold attachment regions, non-coding sequences, splicing signals, transcription termination signals, and polyadenylation signals.
  • An RNA expression cassette typically includes a translation initiation codon (allowing translation initiation), and a sequence encoding one or more proteins.
  • the expression cassette may include translation termination signals, a polyadenosine sequence, internal ribosome entry sites (IRES), and non-coding sequences.
  • the expression cassette promoter can be selected from any of the known promoters isolated from the group consisting of, but not limited to, the human genome, mammalian genomes, microbial genomes, and chimeric sequences. Additionally, artificially constructed sequences can be used that have shown to have promoter activity in the target cell type. Examples of viral promoters that have successfully been used to express transgenes include: human cytomegalovirus immediate early promoter, Rous sarcoma virus, Moloney leukemia virus, and SV40. Examples of mammalian promoters include: elongation factor 1, muscle creatine kinase, actin, desmin, and troponin.
  • Preferential expression in muscle cells can be achieved by using promoters and regulatory elements from muscle-specific genes (e.g., muscle creatine kinase, myosin light chain, desmin, skeletal actin), or by combining transcriptional enhancers from muscle-specific genes with a promoter normally active in many cell types (e.g., the human cytomegalovirus immediate early promoter in combination with the myosin light chain enhancer).
  • muscle-specific genes e.g., muscle creatine kinase, myosin light chain, desmin, skeletal actin
  • transcriptional enhancers from muscle-specific genes e.g., muscle creatine kinase, myosin light chain, desmin, skeletal actin
  • transcriptional enhancers from muscle-specific genes e.g., muscle creatine kinase, myosin light chain, desmin, skeletal actin
  • transcriptional enhancers from muscle-specific genes e.g., muscle creatine kinase, my
  • Muscle specific promoters may be selected from the list comprising: muscle creatine kinase (MCK), myosin light chain, myosin light chain 3F, desmin, alpha-actin, enolase, utrophin, dystrophin, sarcoglycan and other dystrophin-associated glycoprotein promoters.
  • MCK muscle creatine kinase
  • Still other transcription elements that function in muscle include: actin and ⁇ -actin promoters, E-box elements, MEF-2 elements, TEF-1 elements, SRE sites, myogenin enhancer sequences, and viral promoters such as CMV and SV40.
  • Protein refers herein to a linear series of greater than 2 amino acid residues connected one to another as in a polypeptide.
  • a therapeutic effect of the protein in attenuating or preventing the disease state can be accomplished by the protein either staying within the cell, remaining attached to the cell in the membrane, or being secreted and dissociated from the cell where it can enter the general circulation and blood.
  • Secreted proteins that can be therapeutic include hormones, cytokines, interferons, enzymes (e.g.
  • Proteins on the membrane can have a therapeutic effect by providing a receptor for the cell to take up a protein or lipoprotein (e.g., low density lipoprotein receptor).
  • Therapeutic proteins that stay within the cell can be enzymes that clear a circulating toxic metabolite as in phenylketonuria.
  • Intracellular proteins can be part of the cytoskeleton (e.g., actin, dystrophin, myosins, sarcoglycans, and dystroglycans) and thus have a therapeutic effect in cardiomyopathies and musculoskeletal diseases (e.g., Duchenne muscular dystrophy, limb-girdle disease).
  • cardiac contractility e.g., calcium and sodium channels
  • inhibitors of restenosis e.g., nitric oxide synthetase
  • angiogenic factors e.g., angiogenic factor-derived neurotrophic factor-derived neurotrophic factor-derived neurotrophic factor-derived neurotrophic factor-derived neurotrophic factor-derived neurotrophic factor-derived neurotrophic factor-derived neurotrophic factor-derived neurotrophic factor-derived neurotrophic factor, and others.
  • anti-angiogenic factors e.g., anti-angiogenic factors.
  • Proteins are targeted for secretion from cells by the presence of a signal peptide. During transit through the endoplasmic reticulum, the signal peptide is removed by specific proteolytic cleavage. It can be anticipated that secretion of certain proteins can be improved by replacing the endogenous signal peptide with a heterologous signal peptide. This can be accomplished by exchanging the coding regions for the signal peptides in the nucleic acid. For example, the signal from the protein placental alkaline phosphatase (often used in a truncated from as secreted alkaline phosphatase, SEAP) can be used to replace the signal from the protein factor IX. This may result in better secretion of the factor IX protein from muscle cells.
  • SEAP secreted alkaline phosphatase
  • the secreted mature factor IX protein is unaltered and functional.
  • reporter gene/protein systems There are three types of reporter (or marker) gene products that are expressed from reporter genes.
  • the reporter gene/protein systems include:
  • Intracellular gene products included proteins such as luciferase, ⁇ -galactosidase, or chloramphenicol acetyl transferase. Typically, they are enzymes whose enzymatic activity can be easily measured. Intracellular gene products, such as ⁇ -galactosidase or green fluorescent protein, can identify cells expressing the reporter gene. On the basis of the intensity of cellular staining, these reporter gene products also yield qualitative information concerning the amount of foreign protein produced per cell. Secreted gene products such as growth hormone, factor IX, secreted alkaline phosphatase, or alpha1-antitrypsin are useful for determining the amount of a secreted protein that a gene transfer procedure can produce. The reporter gene product can be assayed in a small amount of blood.
  • therapeutic and therapeutic results are defined in this application as a nucleic acid which is transfected into a cell, in vivo, resulting in a gene product (e.g. protein) being expressed in the cell or secreted from the cell.
  • a gene product e.g. protein
  • levels of a gene product, including reporter (marker) gene products are measured which then indicate a reasonable expectation of similar amounts of gene expression by transfecting other nucleic acids.
  • levels of treatment considered beneficial by a person having ordinary skill in the art of gene therapy differ from disease to disease, for example: Hemophilia A and B are caused by deficiencies of the X-linked clotting factors VIII and IX, respectively.
  • Parenchymal cells are the distinguishing cells of a gland or organ contained in and supported by the connective tissue framework.
  • the parenchymal cells typically perform a function that is unique to the particular organ.
  • the term parenchymal often excludes cells that are common to many organs and tissues such as fibroblasts and endothelial cells within blood vessels.
  • the parenchymal cells include hepatocytes, Kupffer cells and the epithelial cells that line the biliary tract and bile ductules.
  • the major constituent of the liver parenchyma are polyhedral hepatocytes (also known as hepatic cells) that presents at least one side to an hepatic sinusoid and opposed sides to a bile canaliculus.
  • Liver cells that are not parenchymal cells include cells within the blood vessels such as the endothelial cells or fibroblast cells.
  • hepatocytes are targeted by injecting the polynucleotide within the tail vein of a rodent such as a mouse.
  • the parenchymal cells include myoblasts, satellite cells, myotubules, and myofibers.
  • the parenchymal cells include the myocardium also known as cardiac muscle fibers or cardiac muscle cells and the cells of the impulse connecting system such as those that constitute the sinoatrial node, atrioventricular node, and atrioventricular bundle.
  • skeletal muscle, cardiac muscle, or diaphragm muscle is targeted by injecting the polynucleotide into the blood vessel supplying or draining the tissue.
  • a polymer is a molecule built up by repetitive bonding together of smaller units called monomers.
  • the term polymer includes both oligomers which have two to about 80 monomers and polymers having more than 80 monomers.
  • the polymer can be linear, branched network, star, comb, or ladder types of polymer.
  • the polymer can be a homopolymer in which a single monomer is used or can be copolymer in which two or more monomers are used. Types of copolymers include alternating, random, block and graft.
  • nucleic acid delivery to cells is the use of nucleic acid-polycations complexes. It was shown that cationic proteins like histones and protamines or synthetic polymers like polylysine, polyarginine, polyornithine, DEAE dextran, polybrene, and polyethylenimine are effective intracellular delivery agents while small polycations like spermine are ineffective.
  • a polycation is a polymer containing a net positive charge, for example poly-L-lysine hydrobromide.
  • the polycation can contain monomer units that are charge positive, charge neutral, or charge negative, however, the net charge of the polymer must be positive.
  • a polycation also can mean a non-polymeric molecule that contains two or more positive charges.
  • a polyanion is a polymer containing a net negative charge, for example polyglutamic acid. The polyanion can contain monomer units that are charge negative, charge neutral, or charge positive, however, the net charge on the polymer must be negative.
  • a polyanion can also mean a non-polymeric molecule that contains two or more negative charges.
  • polyion includes polycation, polyanion, zwitterionic polymers, and neutral polymers that contain equal amounts of anions and cations.
  • zwitterionic refers to the product (salt) of the reaction between an acidic group and a basic group that are part of the same molecule. Salts are ionic compounds that dissociate into cations and anions when dissolved in solution. Salts increase the ionic strength of a solution, and consequently decrease interactions between nucleic acids with other cations.
  • polycations are mixed with polynucleotides for intravascular delivery to a cell.
  • Polycations provide the advantage of allowing attachment of DNA to the target cell surface.
  • the polymer forms a cross-bridge between the polyanionic nucleic acids and the polyanionic surfaces of the cells.
  • the main mechanism of DNA translocation to the intracellular space might be non-specific adsorptive endocytosis which may be more effective then liquid endocytosis or receptor-mediated endocytosis.
  • polycations are a very convenient linker for attaching specific receptors to DNA and as result, DNA-polycation complexes can be targeted to specific cell types.
  • polycations protect DNA in complexes against nuclease degradation. This is important for both extra- and intracellular preservation of DNA.
  • the endocytic step in the intracellular uptake of DNA-polycation complexes is suggested by results in which DNA expression is only obtained by incorporating a mild hypertonic lysis step (either glycerol or DMSO).
  • Gene expression is also enabled or increased by preventing endosome acidification with NH 4 CI or chloroquine.
  • Polyethylenimine which facilitates gene expression without additional treatments probably disrupts endosomal function itself. Disruption of endosomal function has also been accomplished by linking the polycation to endosomal-disruptive agents such as fusion peptides membrane active compounds, or adenoviruses.
  • membrane active compound If the membrane active compound is too active or active at the wrong time, then no transport occurs or transport is associated with cell rupture and thereby cell death.
  • Nature has evolved various strategies to allow for membrane transport of biologically active compounds including membrane fusion and the use membrane active compounds whose activity is modulated such that activity assists transport without toxicity.
  • Many lipid-based transport formulations rely on membrane fusion and some membrane active peptides' activities are modulated by pH.
  • viral coat proteins are often pH-sensitive, inactive at neutral or basic pH and active under the acidic conditions found in the endosome.
  • Polycations also cause DNA condensation.
  • the volume which one DNA molecule occupies in complex with polycations is drastically lower than the volume of a free DNA molecule.
  • the size of DNA/polymer complex may be important for gene delivery in vivo. In terms of intravenous injection, DNA must cross the endothelial barrier and reach the parenchymal cells of interest.
  • liver fenestrae holes in the endothelial barrier
  • increases in pressure and/or permeability can increase the size of the fenestrae.
  • the fenestrae size in other organs is usually less.
  • the size of the DNA complexes is also important for the cellular uptake process. After binding to the target cells the DNA-polycation complex is expected to be taken up by endocytosis.
  • Polymers may incorporate compounds that increase their utility. These groups can be incorporated into monomers prior to polymer formation or attached to the polymer after its formation.
  • the gene transfer enhancing signal (Signal) is defined in this specification as a molecule that modifies the nucleic acid complex and can direct it to a cell location (such as tissue cells) or location in a cell (such as the nucleus) either in culture or in a whole organism. By modifying the cellular or tissue location of the foreign gene, the expression of the foreign gene can be enhanced.
  • the gene transfer enhancing signal can be a protein, peptide, lipid, steroid, sugar, carbohydrate, nucleic acid or synthetic compound.
  • the gene transfer enhancing signals enhance cellular binding to receptors, cytoplasmic transport to the nucleus and nuclear entry or release from endosomes or other intracellular vesicles.
  • Nuclear localizing signals enhance the targeting of the gene into proximity of the nucleus and/or its entry into the nucleus.
  • Such nuclear transport signals can be a protein or a peptide such as the SV40 large T ag NLS or the nucleoplasmin NLS.
  • These nuclear localizing signals interact with a variety of nuclear transport factors such as the NLS receptor (karyopherin alpha) which then interacts with karyopherin beta.
  • the nuclear transport proteins themselves could also function as NLS's since they are targeted to the nuclear pore and nucleus.
  • Signals that enhance release from intracellular compartments can cause DNA release from intracellular compartments such as endosomes (early and late), lysosomes, phagosomes, vesicle, endoplasmic reticulum, Golgi apparatus, trans Golgi network (TGN), and sarcoplasmic reticulum. Release includes movement out of an intracellular compartment into cytoplasm or into an organelle such as the nucleus. Releasing signals include chemicals such as chloroquine, bafilomycin or Brefeldin A1 and the ER-retaining signal (KDEL sequence), viral components such as influenza virus hemagglutinin subunit HA-2 peptides and other types of amphipathic peptides.
  • Cellular receptor signals are any signal that enhances the association of the gene with a cell. This can be accomplished by either increasing the binding of the gene to the cell surface and/or its association with an intracellular compartment, for example: ligands that enhance endocytosis by enhancing binding the cell surface. This includes agents that target to the asialoglycoprotein receptor by using asialoglycoproteins or galactose residues. Other proteins such as insulin, EGF, or transferrin can be used for targeting. Peptides that include the RGD sequence can be used to target many cells. Chemical groups that react with sulfhydryl or disulfide groups on cells can also be used to target many types of cells. Folate and other vitamins can also be used for targeting. Other targeting groups include molecules that interact with membranes such as lipids fatty acids, cholesterol, dansyl compounds, and amphotericin derivatives. In addition viral proteins could be used to bind cells.
  • polynucleotide or nucleic acid
  • Nucleotides are the monomeric units of polynucleotide polymers. Polynucleotides with less than 120 monomeric units are often called oligonucleotides.
  • Natural nucleic acids have a deoxyribose- or ribose-phosphate backbone.
  • An artificial or synthetic polynucleotide is any polynucleotide that is polymerized in vitro or in a cell free system and contains the same or similar bases but may contain a backbone of a type other than the natural ribose-phosphate backbone.
  • backbones include: PNAs (peptide nucleic acids), phosphorothioates, phosphorodiamidates, morpholinos, and other variants of the phosphate backbone of native nucleic acids.
  • Bases include purines and pyrimidines, which further include the natural compounds adenine, thymine, guanine, cytosine, uracil, inosine, and natural analogs. Synthetic derivatives of purines and pyrimidines include, but are not limited to, modifications which place new reactive groups such as, but not limited to, amines, alcohols, thiols, carboxylates, and alkylhalides.
  • base encompasses any of the known base analogs of DNA and RNA including, but not limited to, 4-acetylcytosine, 8-hydroxy-N6-methyladenosine, aziridinylcytosine, pseudoisocytosine, 5-(carboxyhydroxylmethyl)uracil, 5-fluorouracil, 5-bromouracil, 5-carboxymethylaminomethyl-2-thiouracil, 5-carboxy-methylaminomethyluracil, dihydrouracil, inosine, N6-isopentenyladenine, 1-methyladenine, 1-methylpseudo-uracil, 1-methylguanine, 1-methylinosine, 2,2-dimethyl-guanine, 2-methyladenine, 2-methylguanine, 3-methyl-cytosine, 5-methylcytosine, N6-methyladenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxy-amino-methyl-2-thiouracil, ⁇ -D-
  • DNA may be in form of cDNA, in vitro polymerized DNA, plasmid DNA, parts of a plasmid DNA, genetic material derived from a virus, linear DNA, vectors (P1, PAC, BAC, YAC, and artificial chromosomes), expression cassettes, chimeric sequences, recombinant DNA, chromosomal DNA, an oligonucleotide, anti-sense DNA, or derivatives of these groups.
  • RNA may be in the form of oligonucleotide RNA, tRNA (transfer RNA), snRNA (small nuclear RNA), rRNA (ribosomal RNA), mRNA (messenger RNA), in vitro polymerized RNA, recombinant RNA, chimeric sequences, anti-sense RNA, siRNA (small interfering RNA), ribozymes, or derivatives of these groups.
  • An anti-sense polynucleotide is a polynucleotide that interferes with the function of DNA and/or RNA.
  • Antisense polynucleotides include, but are not limited to: morpholinos, 2′-O-methyl polynucleotides, DNA, RNA and the like.
  • SiRNA comprises a double stranded structure typically containing 15-50 base pairs and preferably 21-25 base pairs and having a nucleotide sequence identical or nearly identical to an expressed target gene or RNA within the cell. Interference may result in suppression of expression.
  • the polynucleotide can be a sequence whose presence or expression in a cell alters the expression or function of cellular genes or RNA.
  • DNA and RNA may be single, double, triple, or quadruple stranded. Double, triple, and quadruple stranded polynucleotide may contain both RNA and DNA or other combinations of natural and/or synthetic nucleic acids.
  • a polynucleotide can be delivered to a cell to express an exogenous nucleotide sequence, to inhibit, eliminate, augment, or alter expression of an endogenous nucleotide sequence, or to express a specific physiological characteristic not naturally associated with the cell.
  • Polynucleotides may be coded to express a whole or partial protein, or may be anti-sense.
  • a delivered polynucleotide can stay within the cytoplasm or nucleus apart from the endogenous genetic material.
  • the polymer could recombine (become a part of) the endogenous genetic material.
  • DNA can insert into chromosomal DNA by either homologous or non-homologous recombination.
  • RNA function inhibitor comprises any nucleic acid or nucleic acid analog containing a sequence whose presence or expression in a cell causes the degradation of or inhibits the function or translation of a specific cellular RNA, usually a mRNA, in a sequence-specific manner.
  • An RNA function inhibitor may also inhibit the transcription of a gene into RNA. Inhibition of RNA can thus effectively inhibit expression of a gene from which the RNA is transcribed.
  • RNA function inhibitors are selected from the group comprising: siRNA, interfering RNA or RNAi, dsRNA, RNA Polymerase III transcribed DNAs, ribozymes, and antisense nucleic acid, which may be RNA, DNA, or artificial nucleic acid.
  • SiRNA comprises a double stranded structure typically containing 15-50 base pairs and preferably 21-25 base pairs and having a nucleotide sequence identical or nearly identical to an expressed target gene or RNA within the cell.
  • Antisense polynucleotides include, but are not limited to: morpholinos, 2′-O-methyl polynucleotides, DNA, RNA and the like.
  • RNA polymerase III transcribed DNAs contain promoters, such as the U6 promoter. These DNAs can be transcribed to produce small hairpin RNAs in the cell that can function as siRNA or linear RNAs that can function as antisense RNA.
  • the RNA function inhibitor may be polymerized in vitro, recombinant RNA, contain chimeric sequences, or derivatives of these groups.
  • the RNA function inhibitor may contain ribonucleotides, deoxyribonucleotides, synthetic nucleotides, or any suitable combination such that the target RNA and/or gene is inhibited.
  • these forms of nucleic acid may be single, double, triple, or quadruple stranded.
  • Vectors are polynucleic molecules originating from a virus, a plasmid, or the cell of a higher organism into which another nucleic fragment of appropriate size can be integrated; vectors typically introduce foreign DNA into host cells, where it can be reproduced. Examples are plasmids, cosmids, and yeast artificial chromosomes; vectors are often recombinant molecules containing DNA sequences from several sources.
  • a vector includes a viral vector: for example, adenovirus; DNA; adenoassociated viral vectors (AAV) which are derived from adenoassociated viruses and are smaller than adenoviruses; and retrovirus (any virus in the family Retroviridae that has RNA as its nucleic acid and uses the enzyme reverse transcriptase to copy its genome into the DNA of the host cell's chromosome; examples include VSV G and retroviruses that contain components of lentivirus including HIV type viruses).
  • a viral vector for example, adenovirus
  • DNA adenoassociated viral vectors
  • retrovirus any virus in the family Retroviridae that has RNA as its nucleic acid and uses the enzyme reverse transcriptase to copy its genome into the DNA of the host cell's chromosome
  • retrovirus any virus in the family Retroviridae that has RNA as its nucleic acid and uses the enzyme reverse transcriptase to copy its genome into the DNA of the host cell's chromosome;
  • a vector is used in this specification to mean any DNA molecule which could include associate molecules to transfer DNA sequences into a cell for expression. Examples include naked DNA, non-viral DNA complexes (e.g. DNA plus polymers (cationic or anionic), DNA plus transfection enhancing compounds, and DNA plus amphipathic compounds) and viral particles.
  • naked DNA e.g. DNA plus polymers (cationic or anionic), DNA plus transfection enhancing compounds, and DNA plus amphipathic compounds
  • a non-viral vector is defined as a vector that is not assembled within a eukaryotic cell including non-viral DNA/polymer complexes, DNA with transfection enhancing compounds and DNA+amphipathic compounds.
  • Skin is the external covering of a mammalian body including the epidermis, the dermis, and the subcutaneous tissue.
  • the permeability of the vessel is increased.
  • Efficiency of polynucleotide delivery and expression was increased by increasing the permeability of a blood vessel within the target tissue.
  • Permeability is defined here as the propensity for macromolecules, such as polynucleotides, to move through vessel walls and enter the extravascular space.
  • One measure of permeability is the rate at which macromolecules move through the vessel wall and out of the vessel.
  • Another measure of permeability is the lack of force that resists the movement of polynucleotides being delivered to leave the intravascular space.
  • obstruct in this specification, is to block or inhibit inflow or outflow of blood in a vessel. Rapid injection may be combined with obstructing the outflow to increase permeability.
  • an afferent vessel supplying an organ is rapidly injected and the efferent vessel draining the tissue is ligated transiently.
  • the efferent vessel (also called the venous outflow or tract) draining outflow from the tissue is also partially or totally impeded for a period of time sufficient to allow delivery of a polynucleotide.
  • an efferent is injected and an afferent vessel flow is impeded.
  • the intravascular pressure of a blood vessel is increased by increasing the osmotic pressure within the blood vessel.
  • hypertonic solutions containing salts such as NaCl, sugars or polyols such as mannitol are used.
  • Hypertonic means that the osmolarity of the injection solution is greater than physiologic osmolarity.
  • Isotonic means that the osmolarity of the injection solution is the same as the physiological osmolarity (the tonicity or osmotic pressure of the solution is similar to that of blood).
  • Hypertonic solutions have increased tonicity and osmotic pressure similar to the osmotic pressure of blood and cause cells to shrink.
  • the permeability of the blood vessel can also be increased by a biologically-active molecule.
  • a biologically-active molecule is a protein or a simple chemical such as papaverine or histamine that increases the permeability of the vessel by causing a change in function, activity, or shape of cells within the vessel wall such as the endothelial or smooth muscle cells.
  • biologically-active molecules interact with a specific receptor or enzyme or protein within the vascular cell to change the vessel's permeability.
  • Biologically-active molecules include vascular permeability factor (VPF) which is also known as vascular endothelial growth factor (VEGF).
  • VPF vascular permeability factor
  • VEGF vascular endothelial growth factor
  • Another type of biologically-active molecule can also increase permeability by changing the extracellular connective material. For example, an enzyme could digest the extracellular material and increase the number and size of the holes of the connective material.
  • Another type of biologically-active molecule is a chelator that
  • a non-viral vector along with a polynucleotide is intravascularly injected in a large injection volume.
  • the injection volume is dependent on the size of the animal to be injected and can be from 1.0 to 3.0 ml or greater for small animals (i.e. tail vein injections into mice).
  • the injection volume for rats can be from 6 to 35 ml or greater.
  • the injection volume for primates can be 70 to 200 ml or greater.
  • the injection volumes in terms of ml/body weight can be 0.03 ml/g to 0.1 ml/g or greater.
  • the injection volume can also be related to the target tissue.
  • delivery of a non-viral vector with a polynucleotide to a limb can be aided by injecting a volume greater than 5 ml per rat limb or greater than 70 ml for a primate.
  • the injection volumes in terms of ml/limb muscle are usually within the range of 0.6 to 1.8 ml/g of muscle but can be greater.
  • delivery of a polynucleotide to liver in mice can be aided by injecting the non-viral vector-polynucleotide in an injection volume from 0.6 to 1.8 ml/g of liver or greater.
  • the complex in another preferred embodiment, can be in an injection volume from 0.6 to 1.8 ml/g of limb muscle or anywhere within this range.
  • the injection fluid is injected into a vessel rapidly.
  • the speed of the injection is partially dependent on the volume to be injected, the size of the vessel to be injected into, and the size of the animal.
  • the total injection volume (1-3 mls) can be injected from 5 to 15 seconds into the vascular system of mice.
  • the total injection volume (6-35 mls) can be injected into the vascular system of rats from 20 to 7 seconds.
  • the total injection volume (80-200 mls) can be injected into the vascular system of monkeys from 120 seconds or less.
  • a large injection volume is used and the rate of injection is varied. Injection rates of less than 0.012 ml per gram (animal weight) per second are used in this embodiment. In another embodiment injection rates of less than ml per gram (target tissue weight) per second are used for gene delivery to target organs. In another embodiment injection rates of less than 0.06 ml per gram (target tissue weight) per second are used for gene delivery into limb muscle and other muscles of primates.
  • a prerequisite for gene expression is that once DNA/polymer complexes have entered a cell the polynucleotide must be able to dissociate from the cationic polymer. This may occur within cytoplasmic vesicles (i.e. endosomes), in the cytoplasm, or the nucleus.
  • cytoplasmic vesicles i.e. endosomes
  • Negatively charged polymers can be fashioned in a similar manner, allowing the condensed nucleic acid particle (DNA plus polycation) to be recharged with a cleavable anionic polymer resulting in a particle with a net negative charge that after reduction of disulfide bonds will release the polynucleic acid.
  • the reduction potential of the disulfide bond in the reducible co-monomer can be adjusted by chemically altering the disulfide bonds environment. This will allow the construction of particles whose release characteristics can be tailored so that the polynucleic acid is released at the proper point in the delivery process.
  • a cellular transport step that has importance for gene transfer and drug delivery is that of release from intracellular compartments such as endosomes (early and late), lysosomes, phagosomes, vesicle, endoplasmic reticulum, Golgi apparatus, trans Golgi network (TGN), and sarcoplasmic reticulum. Release includes movement out of an intracellular compartment into cytoplasm or into an organelle such as the nucleus. Chemicals such as chloroquine, bafilomycin or Brefeldin A1 may increase release of molecules into the cytoplasm of a cell. Chloroquine decreases the acidification of the endosomal and lysosomal compartments but also affects other cellular functions.
  • Brefeldin A an isoprenoid fungal metabolite, collapses reversibly the Golgi apparatus into the endoplasmic reticulum and the early endosomal compartment into the trans-Golgi network (TGN) to form tubules.
  • Bafilomycin A1 a macrolide antibiotic is a more specific inhibitor of endosomal acidification and vacuolar type H + -ATPase than chloroquine.
  • the ER-retaining signal (KDEL sequence) has been proposed to enhance delivery to the endoplasmic reticulum and prevent delivery to lysosomes.
  • DNA-polycation particles that form a third layer in the DNA complex and make the particle negatively charged.
  • polyanions that are cleaved in the acid conditions found in the endosome, pH 5-7.
  • cleavage of polymers in the DNA complexes in the endosome assists in endosome disruption and release of DNA into the cytoplasm.
  • cleave a polyion There are two ways to cleave a polyion: cleavage of the polymer backbone resulting in smaller polyions or cleavage of the link between the polymer backbone and the ion containing groups resulting in small ionized molecules and a polymer. In either case, the interaction between the polyion and DNA is broken and the number of molecules in the endosome increases. This causes an osmotic shock to the endosomes and disrupts the endosomes. In the second case, if the polymer backbone is hydrophobic it may interact with the membrane of the endosome. Either effect may disrupt the endosome and thereby assist in release of DNA.
  • cleavable polymers To construct cleavable polymers, one may attach the ions or polyions together with bonds that are inherently labile such as disulfide bonds, diols, diazo bonds, ester bonds, sulfone bonds, acetals, ketals, enol ethers, enol esters, imines, imminiums, and enamines
  • bonds that are inherently labile such as disulfide bonds, diols, diazo bonds, ester bonds, sulfone bonds, acetals, ketals, enol ethers, enol esters, imines, imminiums, and enamines
  • bonds that are inherently labile such as disulfide bonds, diols, diazo bonds, ester bonds, sulfone bonds, acetals, ketals, enol ethers, enol esters, imines, imminiums, and enamines
  • Examples include having carboxylic acid derivatives (acids, esters, amides) and alcohols, thiols, carboxylic acids or amines in the same molecule reacting together to make esters, thiol esters, acid anhydrides or amides.
  • the present invention additionally provides for the use of polymers containing silicon-nitrogen (silazanes) linkages (either in the main chain of the polymer or in a side chain of the polymer) that are susceptible to hydrolysis.
  • Hydrolysis of a silazane leads to the formation of a silanol and an amine.
  • Silazanes are inherently more susceptible to hydrolysis than is the silicon-oxygen-carbon linkage; however, the rate of hydrolysis is increased under acidic conditions.
  • the substitution on both the silicon atom and the amine can affect the rate of hydrolysis due to steric and electronic effects. This allows for the possibility of tuning the rate of hydrolysis of the silizane by changing the substitution on either the silicon or the amine to facilitate the desired affect.
  • ester acids and amide acids that are labile in acidic environments (pH less than 7, greater than 4) to form an alcohol and amine and an anhydride are use in a variety of molecules and polymers that include peptides, lipids, and multimolecular associations such as liposomes.
  • ketals that are labile in acidic environments (pH less than 7, greater than 4) to form a diol and a ketone are use in a variety of molecules and polymers that include peptides, lipids, and liposomes.
  • acetals that are labile in acidic environments (pH less than 7, greater than 4) to form a diol and an aldehyde are use in a variety of molecules and polymers that include peptides, lipids, and liposomes.
  • enols that are labile in acidic environments (pH less than 7, greater than 4) to form a ketone and an alcohol are use in a variety of molecules and polymers that include peptides, lipids, and liposomes.
  • iminiums that are labile in acidic environments (pH less than 7, greater than 4) to form an amine and an aldehyde or a ketone are use in a variety of molecules and polymers that include peptides, lipids, and liposomes.
  • peptides and polypeptides are modified by an anhydride.
  • the amine (lysine), alcohol (serine, threonine, tyrosine), and thiol (cysteine) groups of the peptides are modified by an anhydride to produce an amide, ester or thioester acid.
  • the amide, ester, or thioester is cleaved displaying the original amine, alcohol, or thiol group and the anhydride.
  • a variety of endosomolytic and amphipathic peptides can be used in this embodiment.
  • a positively-charged amphipathic/endosomolytic peptide is converted to a negatively-charged peptide by reaction with the anhydrides to form the amide acids and this compound is then complexed with a polycation-condensed nucleic acid. After entry into the endosomes, the amide acid is cleaved and the peptide becomes positively charged and is no longer complexed with the polycation-condensed nucleic acid and becomes amphipathic and endosomolytic.
  • the peptides contains tyrosines and lysines.
  • the hydrophobic part of the peptide (after cleavage of the ester acid) is at one end of the peptide and the hydrophilic part (e.g. negatively charged after cleavage) is at another end.
  • the hydrophobic part could be modified with a dimethylmaleic anhydride and the hydrophilic part could be modified with a citranconyl anhydride. Since the dimethylmaleyl group is cleaved more rapidly than the citraconyl group, the hydrophobic part forms first.
  • the hydrophilic part forms alpha helixes or coil-coil structures.
  • the ester, amide or thioester acid is complexed with lipids and liposomes so that in acidic environments the lipids are modified and the liposome becomes disrupted, fusogenic or endosomolytic.
  • the lipid diacylglycerol is reacted with an anhydride to form an ester acid. After acidification in an intracellular vesicle the diacylglycerol reforms and is very lipid bilayer disruptive and fusogenic.
  • cationic polymers such as histone (H1, H2a, H2b, H3, H4, H5), HMG proteins, poly-L-lysine, polyethylenimine, protamine, and poly-histidine are used to compact polynucleic acids to help facilitate gene delivery in vitro and in vivo.
  • H1, H2a, H2b, H3, H4, H5 HMG proteins
  • poly-L-lysine polyethylenimine
  • protamine protamine
  • poly-histidine poly-histidine
  • Tail vein injection process having non-cleavable cationic polymer/DNA complexes we found that gene expression is most efficient when the overall net charge of the complexes are negative (DNA negative charge>polycation positive charge).
  • Tail vein injections using cationic polymers commonly used for DNA condensation and in vitro gene delivery revealed that high gene expression occurred when the net charge of the complexes were negative.
  • angiogenesis in this specification is defined as any formation of new blood vessels.
  • Angiogenesis may also refer to the sprouting of new blood vessels (endothelium-lined channels such as capillaries) from pre-existing vessels as a result of proliferation and migration of endothelial cells.
  • arteriogenesis The maturation or enlargement of vessels via recruitment of smooth muscle cells, i.e. the formation of collateral arteries from pre-existing arterioles, is termed arteriogenesis.
  • Vasculogenesis refers to the in situ formation of blood vessels from angioblasts and endothelial precursor cells (EPCs).
  • EPCs endothelial precursor cells
  • An anastomosis is a connection between two blood vessels. The formation of anastomoses can be important for restoring blood flow to ischemic tissue.
  • angiogenesis encompasses arteriogenesis, vasculogenesis, anastomosis formation, and revascularization.
  • Angiogenesis is regulated by soluble secreted factors, cell surface receptors and transcription factors.
  • Secreted factors include cytokines, chemokines, and growth factors that affect endothelial cells, smooth muscle cells, monocytes, leukocytes, and precursor cells.
  • Such factors include: vascular endothelial growth factors, fibroblast growth factors, hepatocyte growth factors, angiopoietin 1 (Ang-1), angiopoietin 2 (Ang-2), Platelet derived growth factors (PDFGs), granulocyte macrophage-colony stimulating factor, insulin-like growth factor-1 (IGF-1), IGF-2, early growth response factor-1 (EGR-1), and human tissue kallikrein (HK).
  • genes that encode angiogenic factors to cells in vivo provides an attractive alternative to repetitive injections of protein for the treatment of vascular insufficiency or occlusions.
  • Genes that encode angiogenic factors can be targeted to cells in the affected area, thereby limiting deleterious effects associated with delivering angiogenic factors throughout the body.
  • genes for angiogenic factors can be delivered to muscle cells in vivo, including skeletal and cardiac muscle cells. Expression of the gene and secretion of the gene product then induces angiogenesis and improves collateral blood flow in the targeted tissue. The improved blood flow can both improve muscle tissue function and relieve pain associated with vascular diseases.
  • the intra-arterial procedure requires that blood flow be impeded for substantially less than the couple of hours of ischemia required for tissue damage.
  • a common anesthesia for human limb surgery e.g., carpal tunnel repair
  • the minimal elevations of muscle-derived enzymes in the serum also argues against any consequential muscle damage.
  • the intravascular pressure can be damaging to the arteries. We have observed minimal intimal changes in the arteries that are presumed to be transient and without consequence. Nonetheless, this minimal arterial damage may be prevented by better controlling the intravascular pressure.
  • the procedure requires relatively large amounts of pDNA to be administered. This has not been associated with any toxic effects in rodents or monkeys. Given that the tourniquet delays pDNA distribution outside of the limb and the intravascular pDNA is rapidly degraded by circulating DNases, pDNA toxicity is unlikely. In addition, the cost for producing clinical grade pDNA is considerably less expensive than viral vectors and does not represent an obstacle to its clinical use.
  • a sphygmomanometer cuff surrounding the arm or leg proximal to the injection site.
  • the catheterized vessels were injected with 30 ml of normal saline containing 5 mg papaverine (Sigma Co.). Five min. later, a saline solution containing 100 ⁇ g pDNA/ml solution was rapidly injected within 30 to 45 sec.
  • the volume of each injection was 75 ml and 90 ml in the first two animals and 120 ml thereafter. The injection volume was 180 ml for the lower legs.
  • the DNA solutions were injected using a nitrogen-pressurized cylinder. Two min after injection, the catheters were removed and the sphygmomanometer deflated.
  • the pCI-Luc + (Promega, Madison, Wis.) and pCI-LacZ plasmids express a cytoplasmic luciferase and the Escherichia coli LacZ, respectively, from the human cytomegalovirus (CMV) immediate-early promoter.
  • CMV human cytomegalovirus
  • the pCI vector (Promega) also contains an SV40 polyadenylation signal.
  • pMI-Luc + was constructed by replacing the CMV promoter in pCI-Luc + with a 3300-bp murine muscle creatine kinase promoter.
  • the vector pEBFP-N1 expresses a nuclear-localizing, blue-shifted green fluorescent protein (GFP) from the CMV promoter (Clontech, Palo Alto, Calif.).
  • GFP nuclear-localizing, blue-shifted green fluorescent protein
  • Luciferase assays were performed on muscle biopsies, entire muscles and various tissues as previously reported.
  • muscle samples were taken from the proximal, middle, and distal positions of each muscle, cut into small pieces, frozen in cold isopentane, and stored at ⁇ 80° C. Muscle pieces were randomly chosen from each muscle sample (for every position) and 10 ⁇ m-thick cryostat sections were made. Every tenth section, for a total of 20 sections, was stained and analyzed.
  • the sections were incubated in X-gal staining solution (5 mM potassium ferricyanide, 5 mM potassium ferrocyanide, 1 mM magnesium chloride, 1 mM X-gal in 0.1 M PBS, pH 7.6) for 4-8 hours at room temperature and counterstained with hematoxylin and eosin. Three sections were selected randomly from the 20 sections of each position (usually the 4th, 11th and 17th sections, but an adjacent section was used if these sections were not intact). As previously described, the number of ⁇ -galactosidase-positive and total cells were determined within a cross area in each section by moving the counter grid from the top edge of the section to the bottom and from the left edge to the right.
  • X-gal staining solution 5 mM potassium ferricyanide, 5 mM potassium ferrocyanide, 1 mM magnesium chloride, 1 mM X-gal in 0.1 M PBS, pH 7.6
  • the percentage of ⁇ -galactosidase-positive cells for each muscle was gotten from the result of positive number divided by total cell number.
  • a weighted average for the percent of transfected cells for each extremity muscle was determined as follows: ( ⁇ Ai*Mi)/M where Ai is percent of transfected cells for one muscle, Mi—weight of that muscle and M—whole weight of all muscles.
  • Luciferase and ⁇ -galactosidase expression Seven rhesus macaque (6-20 years old) received pDNA injections into their limb arteries. All seven monkeys tolerated the procedure well and had full function of their arms, hands, legs and feet following the procedure. In particular, this indicates lack of damage to the radial nerve, which could have been sensitive to the inflated sphygmomanometer surrounding the upper arm. Swelling in the target limbs, a putative correlate of successful gene transfer, was noted afterwards but completely subsided by the next day. When the monkeys awakened from the anesthesia 15 to 30 min after the procedure, they did not appear to be in any discomfort beyond that of normal surgical recovery. Occasionally, the skin in the target limb had some spots of hemorrhage that resolved within several days.
  • Toxicity Serum chemistries and histologic analyses were performed to determine if the procedure caused any adverse effects in the monkeys.
  • CK creatine phosphate kinase
  • AST aspartate aminotransferase
  • LDH lactate dehydrogenase
  • GTT ⁇ -glutamyltransferase
  • alkaline phosphatase hematological assays (hematocrit and RBC indices, platelets), serum electrolytes (Na, Cl, K), serum minerals (calcium, phosphate, iron), serum proteins (albumin, total protein), serum lipids (cholesterol, triglycerides), renal indices (urea, creatinine), and bilirubin were unaffected.
  • Total WBC increased within the typical range post-surgery.
  • Limb muscles were obtained 14 to 16 days after intraarterial injection and examined histologically. The vast majority of muscle tissue was well preserved and did not show any sign of pathology. In a few sections, mononuclear cells were noted surrounding ⁇ -galactosidase positive myofibers, some of which were undergoing degeneration. Immunostaining for CD-markers indicated that the majority of infiltrating cells were CD3-positive (T lymphocytes) with only a few B cells.
  • Muscle luciferase expression was measured at several time points following intravascular delivery of the luciferase gene under control of either the CMV promoter (pCI-Luc + ) or MCK promoter (pMI-Luc + ) into: a) untreated rats, b) rats continuously immunosuppressed (treated with 2.5 mg/kg of FK506 orally and 1 mg/kg dexamethasone subcutaneously one day prior to, one hour prior to and every day thereafter with FK506) or c) transiently immunosuppressed (treated with 10 mg/kg of FK506 orally and 1 mg/kg dexamethasone subcutaneously one day prior to, one hour prior to and one day after intraarterial delivery of pDNA) (Table 2).
  • Rhodamine-labeled pDNA was injected into the femoral artery of rats under various conditions in order to explore the uptake mechanism in muscle as was done for liver.
  • Rh-pDNA Rhodamine-labeled pDNA
  • FIG. 3A presents a rare field when some DNA can be seen between muscle cells.
  • Rh-pDNA was detected throughout all the muscle ( FIGS. 3B and 3 C). At 5 min after injection, examination of tissue sections indicated that the majority of the Rh-pDNA was surrounding the muscle cells and there was no intracellular staining ( FIG.
  • a plasmid DNA (pCI-hF9) expressing the human factor IX gene (cDNA) under transcriptional control of the human cytomegalovirus promoter was delivered to rat hind limb skeletal muscle. A midline abdominal incision was made and skin flaps were folded away with clamps to expose target area. Intestines were moved to visualize the iliac veins and arteries. Microvessel clips were placed on the external iliac, caudal epigastric, internal iliac, deferent duct, and gluteal arteries and veins to block both outflow and inflow of the blood to the leg.
  • An efflux enhancer solution (0.5 mg papaverine in 3 ml saline) was injected into the external iliac artery though a 25 g needle, followed by the pDNA containing solution (500 ⁇ g in 10 ml Ringer's) 5 minutes later.
  • the pDNA solution was injected in approximately 10 seconds.
  • the microvessel clips were removed 2 minutes after the injection, and the peritoneum and skin were closed using sutures.
  • the rats were immunosuppressed by treatment with 10 mg/kg of FK506 orally and 1 mg/kg dexamethasone subcutaneously one day prior to, one hour prior to, and one day after plasmid DNA delivery
  • the rats were sacrificed after 3 weeks, at which time the hind limb skeletal muscles were removed and homogenized in a total volume of 60 ml.
  • Human factor IX levels in the rat sera were determined using an ELISA and compared to normal human serum. Expression levels in 3 rats were 1400, 1000, and 1150 ng/ml extract, respectively. Therefore, the total amount of human factor IX present in the rat muscle tissue three weeks after pDNA delivery was approximately 70 ⁇ g.
  • a plasmid DNA expression vector (pMIR54) was constructed in which the secreted alkaline phosphatase (SEAP) gene (obtained from plasmid pSEAP-2 basic, Clontech) is under transcriptional control of the human cytomegalovirus promoter.
  • SEAP secreted alkaline phosphatase
  • a solution of 500 ⁇ g pMIR54 in 10 ml Ringer's was injected into the iliac artery of Sprague Dawley rats as described in Examples above. The rats were immunosuppressed by treatment with 2.5 mg/kg of FK506 orally and 1 mg/kg dexamethasone subcutaneously one day prior to, one hour prior to plasmid DNA delivery.
  • SEAP expression was determined using a chemiluminescent assay (Tropix) and compared to a standard curve. TABLE 3 SEAP expression in rat muscle cells following DNA delivery. SEAP expression (ng SEAP per ml serum) Day 7 Day 14 Rat 2889 2,301 1,407 Rat 2992 3,735 2,942
  • Intravascularly-administered plasmid DNA is expressed efficiently in multiple muscle groups when blood flow is impeded using an external tourniquet.
  • ICR or mdx mice ⁇ 30 gram, were anesthetized by intramuscular injection of ketamine (80-100 mg/kg) and xylazine (2 mg/kg). Metofane was added through inhalation if necessary during the procedure. A median incision was made from the upper third of abdomen to the caudal edge of the abdominal and the right caudal part of abdominal cavity was exposed using retractors. The tissue in front of the right external iliac artery was cleaned by forceps and a cotton tipped applicator.
  • the arteries and veins to be clamped were separated from surrounding tissue and the caudal epigastric artery and vein, internal iliac arteries and vein, gluteal artery and vein, the vessels of deferent duct and external iliac artery and vein were clamped.
  • a 0.6 ml of papaverine solution (containing 0.1 mg of papaverine) was injected into external iliac artery distal to the clamp.
  • 2.5-3 ml of DNA solution containing 100 ⁇ g plasmid DNA was injected into the external iliac artery distal to the clamp with pressure 5 minutes post papaverine injection.
  • a piece of gelfoam was put on the injection site before withdrawal of the needle and pressure was kept on the gelfoam to prevent bleeding.
  • the clamps are taken off 2 minutes after injection and the abdominal cavity was closed by suturing.
  • Muscle samples were taken 7-10 days after injection and 6 ⁇ m thickness cryostat sections were made. Endogenous peroxidase activities were blocked by incubating the sections in 0.3% hydrogen peroxide in PBS for 5-10 min after the sections were mounted on slides and dried. The sections were rinsed twice with PBS (2 min ⁇ 2) followed by Avidin/Biotin blocking by using Vector Avidin/Biotin Blocking Kit (Cat. No. SP-2001). The following steps were done according to the procedure of Vector M.O.M Immunodetection Kit (Cat. No. PK-2200). The immunofluorescent staining for human dystrophin in mouse muscle was done following the procedure of Vector M.O.M Immunodetection Kit (Cat. No. FMK-2201).
  • FIG. 4A shows ⁇ -galactosidase expression in mdx dystrophic mouse.
  • FIGS. 4 B and C show human dystrophin expression in leg skeletal muscle in mdx and normal mouse, respectively.
  • TABLE 5 Luciferase expression in Mdx mouse. total expression of whole leg expression of per gram muscle animal # (ng luciferase) (ng luciferase/gram muscle) 134 1691 1271 135 1738 1307 137 1248 1177 138 869 643 140 1641 1357 141 881 663 average 1345 1070
  • Intravenous delivery of the therapeutically relevant dystrophin gene to limb muscle cells in mouse A clinically-relevant example is provided by the intravenous delivery of the human dystrophin gene into the mdx4cv mouse model for Duchenne muscular dystrophy, the dystrophin-negative strain B6Ros.Cg-DMDmdx-4Cv (Jackson laboratory).
  • the ability to perform the intravenous procedure in mouse models enhances its utility as a research tool.
  • brachial vein was clamped. Blood circulation of the forelimb was further inhibited by using a tourniquet placed around the upper limb up to the elbow (10 minutes maximum). For whole hind leg injections, an incision was made through the midline of the abdomen one inch below the umbilicus to the pubis. Connective tissue was separated to expose the common iliac artery and vein, external iliac artery and vein, internal iliac artery and vein, inferior epigastric artery and vein, superficial epigastric artery and vein, and the superficial iliac circumflex artery and vein. Clamps were placed on the inferior epigastric artery and vein, superficial epigastric artery and vein, and the superficial iliac circumflex artery and vein.
  • a catheter (F5) was placed into the distal part of the iliac artery to the femoral artery and secured by ligation at the beginning of the femoral artery. Clamps are then placed on the external iliac vein, internal iliac artery and vein, and the common iliac artery and vein.
  • a 17% papaverine/saline solution was injected to increase vessel dilation (10-50 ml depending on animal size). After 5 minutes a plasmid DNA/saline solution was injected at moderately increased pressure using a nitrogen-pressurized cylinder set at 65 psi. For the forelimbs, the injection volume was 50-200 ml. For whole leg injections, the injection volume was 60-500 ml. Injection rates varied from 20 s to 120 s. Two min after injection, the clamps and tourniquet were released and the catheters were removed.
  • FIG. 6A illustrates the distribution of luciferase expression in normal dog.
  • 6B illustrates the distribution of luciferase expression in the dystrophic dog model. TABLE 6 Luciferase expression after of delivery pCI-Luc polynucleotide in dog skeletal muscle cells. Numbers given in pg Luciferase per mg total protein.
  • PEI/DNA, Histone H1/DNA, and MC66/DNA Particles to Multiple Skeletal Muscles in Rat Via a Single Injection into a Blood Vessel.
  • PEI/DNA and histone H1/DNA particles were injected into rat leg muscle by a single intra-arterial injection into the external iliac (Budker et al. 1998).
  • MC66/DNA particles 500 ⁇ g pDNA (500 ⁇ l) was mixed with M66 copolymer at a 1:3 wt:wt ratio in 500 ⁇ l saline. Complexes were then diluted in Ringers solution to total volume of 10 mls.
  • DNA/PEI particles (1:0.5 charge ratio) Total Total Muscle Group
  • Luciferase muscle group 1 (upper leg anterior) 3.50 ⁇ 10 9 0.180 ⁇ g muscle group 2 (upper leg posterior) 3.96 ⁇ 10 9 0.202 ⁇ g muscle group 3 (upper leg medial) 7.20 ⁇ 10 9 0.368 ⁇ g muscle group 4 (lower leg posterior) 9.90 ⁇ 10 9 0.505 ⁇ g muscle group 5 (lower leg anterior) 9.47 ⁇ 10 8 0.048 ⁇ g muscle group 6 (foot) 6.72 ⁇ 10 6 0.0003 ⁇ g
  • DNA delivery was performed via brachial artery with blood flow blocked by a sphygmomanometer cuff proximately to the injection site.
  • Left arm was transfected with VEGF, while right arm was transfected with EPO.
  • the Sartorious muscle from left leg was used as non-injected control.
  • a male Rhesus monkey weighing 14 kg was used for these injections.
  • the animal was anesthetized with Ketamine (10-15 mg/kg).
  • a modified pediatric blood pressure cuff was positioned on the upper arm.
  • the brachial artery was cannulated with a 4 F angiography catheter.
  • the catheter was advanced so that the tip was positioned just below the blood pressure cuff.
  • the blood pressure cuff was inflated so that the cuff pressure was at least 20 mmHg higher than the systolic blood pressure.
  • papaverine (5 mg in 30 ml of saline) was injected by hand ( ⁇ 8 to 10 seconds).
  • the pDNA solution was delivered rapidly with a high volume injection system.
  • 10 mg of pDNA was added to 170 ml of saline and injected at a rate of 6.8 ml per second.
  • 10 mg of pDNA was added to 150 ml of saline, and injected at a rate of 5.4 ml per second.
  • VEGF plasmid delivery was performed using a standard protocol for paraffin sections. Briefly: four microns paraffin sections were deparaffinized and re-hydrated. Antigen retrieval was performed with DAKO Target Retrieval Solution (DAKO Corporation, Carpinteria Calif.) for 20 min at 97° C. To reduce non-specific binding the section were incubated in PBS containing 1% (wt/vol) BSA for 20 min at RT. Primary antibody 1:30 in PBS/BSA were applied for 30 min at RT.
  • CD31 antibody were visualized with donkey anti-mouse Cy3-conjugated IgG, 1:400 (Jackson Immunoresearch Lab, West Grove Pa.) for 1 h at RT. ToPro-3 (Molecular Probes Inc.) was used for nuclei staining; 1:70,000 dilution incubated for 15 min at RT. Sections were mounted with Vectashield non-fluorescent mounting medium and examined under confocal Zeiss LSM 510 microscope (Carl Zeiss, Goettingen, Germany). Images were collected randomly under 400 ⁇ magnification, each image representing 0.106 sq mm. Because muscle fibers and red blood cells have an autofluorescence in FITC channel we use a 488 nm laser to visualize these structures.
  • Morphometry analysis Coded mages were opened in Adobe Photoshop 5.5 having image size 7 ⁇ 7 inches in 1 ⁇ 7 inches window, and a grid with rulers was overlaid. The number of muscle fibers, CD31 positive cells and total nuclei was counted in all 7 image's strips consecutively, without any knowledge of experimental design. T-Test for Two-Sample Unequal Variances was used for statistical analysis.
  • Results of the rat injections are in relative light units (RLUs) and ⁇ g of luciferase produced.
  • RLUs relative light units
  • 10 ⁇ l of cell lysate were assayed using an EG&G Berthold LB9507 luminometer and total muscle RLUs were determined by multiplying by the appropriate dilution factor.
  • ng luciferase RLUs ⁇ 5.1 ⁇ 10 ⁇ 8 ).
  • the guide wire was directed into the inferior phrenic vein.
  • the catheter position in the inferior phrenic vein was checked by injecting iodine.
  • the balloon was inflated to block blood flow through the inferior phrenic vein.
  • 20 ml 0.017% papaverine in normal saline was injected.
  • 40 ml of DNA solution (3 mg) was injected under elevated pressure (65 sec injection time). 2 min after DNA injection, the balloon was released and the catheter was removed.
  • the animal was sacrificed and the diaphragm was taken for luciferase assay 7 days after the procedure. TABLE 8 Luciferase expression in diaphragm from monkey sacrificed 7 days after injection of pCI-Luc + .
  • M-methyl-L-arginine (L-NMMA) is a competitive inhibitor of nitric oxide synthetase.
  • pCI-Luc was injected into the iliac artery of rat following a short pre-treatment with L-NMMA.
  • a 4 cm long abdominal midline excision was performed in 150-200 g, adult Sprague-Dawley rats anesthetized with 80 mg/mg ketamine and 40 mg/kg xylazine.
  • Microvessel clips were placed on external iliac, caudal epigastric, internal iliac and deferent duct arteries and veins to block both outflow and inflow of the blood to the leg.

Abstract

A process is described for the delivery of a therapeutic polynucleotide to limb muscle tissue suffering from or potentially suffering from Muscular Dystrophy. The polynucleotide is inserted into a mammalian limb vessel such as an artery. Delivery efficiency and distribution is enhanced by combining injection of a solution containing the polynucleotide with the use of an externally applied cuff.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation-in-part of application Ser. No. 10/429,478, filed May 5, 2003, pending, a continuation-in-part of application Ser. No. 10/339,934, filed Jan. 10, 2003, pending, a continuation-in-part of application Ser. No. 09/707,000, filed Nov. 6, 2000, pending, and a continuation-in-part of application Ser. No. 09/707,117, filed Nov. 6, 2000, pending, application Ser. Nos. 09/707,000 and 09/707,117 claims the benefit of U.S. Provisional Application No. 60/163,719, filed Nov. 5, 1999.
  • FIELD OF THE INVENTION
  • The invention relates to devices and processes for use in biological systems. More particularly, processes that provide for the functional distribution of genetic material to mammalian cells are described.
  • BACKGROUND OF THE INVENTION
  • Biotechnology includes the delivery of genetic information to a cell to express an exogenous nucleotide sequence, to inhibit, eliminate, augment, or alter expression of an endogenous nucleotide sequence, or to express a specific physiological characteristic not naturally associated with the cell. Polynucleotides may be coded to express a whole or partial protein, or may be anti-sense, or non-viral DNA, or recombine with chromosomal DNA.
  • A basic challenge for biotechnology and thus its subpart, gene therapy, is to develop approaches for delivering genetic information to cells of a patient in a way that is efficient and safe. This problem of drug delivery, where the genetic material is a drug, is particularly challenging. If genetic material are appropriately delivered they can potentially enhance a patient's health and, in some instances, lead to a cure. Therefore, a primary focus of gene therapy is based on strategies for delivering genetic material in the form of nucleic acids. After delivery strategies are developed they may be sold commercially since they are then useful for developing drugs.
  • Delivery of a nucleic acid means to transfer a nucleic acid from a container outside a mammal to near or within the outer cell membrane of a cell in the mammal. The term transfection is used herein, in general, as a substitute for the term delivery, or, more specifically, the transfer of a nucleic acid from directly outside a cell membrane to within the cell membrane. The transferred (or transfected) nucleic acid may contain an expression cassette. If the nucleic acid is a primary RNA transcript that is processed into messenger RNA, a ribosome translates the messenger RNA to produce a protein within the cytoplasm. If the nucleic acid is a DNA, it enters the nucleus where it is transcribed into a messenger RNA that is transported into the cytoplasm where it is translated into a protein. Therefore if a nucleic acid expresses its cognate protein, then it must have entered a cell. A protein may subsequently be degraded into peptides, which may be presented to the immune system.
  • It was first observed that the in vivo injection of plasmid DNA into muscle enabled the expression of foreign genes in the muscle (Wolff 1990). Since that report, several other studies have reported the ability for foreign gene expression following the direct injection of DNA into the parenchyma of other tissues. Naked DNA was expressed following its injection into cardiac muscle (Acsadi 1991).
  • The muscular dystrophies (MD) are a heterogeneous group of mostly inherited disorders characterized by progressive muscle wasting and weakness which eventually leads to death. In most in not all forms of MD, the disease is associated with either a non-functioning or malfunctioning protein due to the presence of a mutant or deleted gene (Hartigan-O'Connor 2000). Because of the nature of these diseases, few traditional treatments are available. However, because the genes and protein products that are responsible for most of the dystrophies have been identified, delivery of corrective genes offers a promising treatment.
  • Several attributes of striated muscle cells make genetic repair feasible. First, myofibers have a long life span, facilitating long term persistence of delivered genes. Second, for DMD and MCDM, it has been shown that gene replacement in striated muscle alone can alleviate the major features of the disease (Cox 1993, Kuang 1998). Third, dystrophin positive fibers may possess a survival advantage over dystrophin negative fibers, suggesting that only a portion of the fibers need to receive the correcting polynucleotide (Morgan 1993). Finally, only 20% of the normal level of dystrophin is required to be asymptomatic. Thus, low level dystrophin expression in a majority of muscle fibers may be sufficient for elimination of symptoms (Phelps 1995).
  • SUMMARY OF THE INVENTION
  • In a preferred embodiment, a process is described for delivering a polynucleotide into a parenchymal cell of a mammal, comprising making a polynucleotide such as a nucleic acid, inserting the polynucleotide into a mammalian vessel, such as a blood vessel, increasing the permeability of the vessel, and delivering the polynucleotide to the parenchymal cell thereby altering endogenous properties of the cell. Increasing the permeability of the vessel consists of increasing pressure against vessel walls and/or inhibiting the flow of fluid through the vessel.
  • In another preferred embodiment, an in vivo process for delivering a polynucleotide to a parenchymal cell of a mammal is described. The polynucleotide is inserted into a blood vessel and interior blood flow is impeded by external application of compression and the polynucleotide is delivered to the parenchymal cell. The polynucleotide may consist of naked DNA, a viral particle/vector, a non-viral vector or may be a blocking polynucleotide for preventing gene expression. The parenchymal cell may consist of a muscle cell, such as a limb (leg or arm) muscle cell.
  • The process includes externally impeding interior blood flow by externally applying pressure to interior blood vessels such as compressing mammalian skin by applying a tourniquet over the skin. Compressing mammalian skin also includes applying a cuff over the skin such as a sphygmomanometer.
  • In another preferred embodiment, an in vivo process for delivering a polynucleotide to a mammalian cell consists of inserting the polynucleotide into a blood vessel and applying pressure to one or more blood vessels. The pressure is applied externally to mammalian skin and the polynucleotide is delivered to the mammalian cell. However, it is important that the function of the mammal's limbs is not permanently impaired using this process. The process especially consists of a polynucleotide delivered to non-vascular (not of the smooth muscle cells surrounding a vessel) parenchymal cells.
  • In yet another preferred embodiment, a device for applying pressure to mammalian skin for in vivo delivery of a polynucleotide to a mammalian cell is described. The device consists of a cuff, as defined in this specification, applied to mammalian skin to impede fluid flow through the vessel thereby increasing delivery efficiency of the polynucleotide to the mammalian cell.
  • In a preferred embodiment it may be preferential to immunosuppress the host receiving the nucleic acid. Immunosuppression can be of long term or for a short duration. For short term duration, the immunosuppression is preferably supplied around the time of nucleic acid delivery. Immunosuppression can be accomplished by treatment with (combinations of) immunosuppressive drugs like cyclosporin A, ProGraf (FK506), corticosteroids, deoxyspergualin, and dexamethasone. Other methods include blocking of immune cell activation pathways, for instance by treatment with (or expression of) an antibody directed against CTLA4; redirection of activated immune cells by treatment with (ore expression of) chemokines such as MIP-1a, MCP-1 and RANTES; and treatment with immunotoxins, such as a conjugate between anti-CD3 antibody and diphtheria toxin.
  • In a preferred embodiment, the process may be used to deliver a therapeutic polynucleotide to a muscle cell for the treatment of vascular disease or occlusion. The delivered polynucleotide can express a protein or peptide that stimulates angiogenesis, vasculogenesis, arteriogenesis, or anastomoses to improve blood flow to a tissue. The gene may be selected from the list comprising: VEGF, VEGF II, VEGF-B, VEGF-C, VEGF-D, VEGF-E, VEGF121, VEGF138, VEGF145, VEGF165, VEGF189, VEGF206, hypoxia inducible factor 1α (HIF 1α), endothelial NO synthase (eNOS), iNOS, VEFGR-1 (Flt1), VEGFR-2 (KDR/Flk1), VEGFR-3 (Flt4), neuropilin-1, ICAM-1, factors (chemokines and cytokines) that stimulate smooth muscle cell, monocyte, or leukocyte migration, anti-apoptotic peptides and proteins, fibroblast growth factors (FGF), FGF-1, FGF-1b, FGF-1c, FGF-2, FGF-2b, FGF-2c, FGF-3, FGF-3b, FGF-3c, FGF-4, FGF-5, FGF-7, FGF-9, acidic FGF, basic FGF, hepatocyte growth factor (HGF), angiopoietin 1 (Ang-1), angiopoietin 2 (Ang-2), Platelet derived growth factors (PDFGs), PDGF-BB, monocyte chemotactic protein-1, granulocyte macrophage-colony stimulating factor, insulin-like growth factor-1 (IGF-1), IGF-2, early growth response factor-1 (EGR-1), ETS-1, human tissue kallikrein (HK), matrix metalloproteinase, chymase, urokinase-type plasminogen activator and heparinase. The protein or peptide may be secreted or stay within the cell. For proteins and peptides that are secreted, the gene may contain a sequence that codes for a signal peptide. The delivered polynucleotide can also suppress or inhibit expression of an endogenous gene or gene product that inhibits angiogenesis, vasculogenesis, arteriogenesis or anastomosis formation. Multiple polynucleotides or polynucleotides containing more that one therapeutic gene may be delivered using the described process. The gene or genes can be delivered to stimulate vessel development, stimulate collateral vessel development, promote peripheral vascular development, improve blood flow in a muscle tissue, or to improve abnormal cardiac function. The gene or genes can also be delivered to treat peripheral circulatory disorders, myocardial disease, myocardial ischemia, limb ischemia, arterial occlusive disease, peripheral arterial occlusive disease, vascular insufficiency, vasculopathy, arteriosclerosis obliterans, thromboangiitis obliterans, atherosclerosis, aortitis syndrome, Behcet's disease, collagenosis, ischemia associated with diabetes, claudication, intermittent claudication, Raynaud disease, cardiomyopathy or cardiac hypertrophy. The polynucleotide can be delivered to a muscle cell that is suffering from ischemia or a normal muscle cell. The muscle cell can be a cardiac cell or a skeletal muscle cell. A preferred skeletal muscle cell is a limb skeletal muscle cell. The polynucleotides can also be delivered to cells in a tissue that is at risk of suffering from ischemia or a vascular disease or disorder.
  • In a preferred embodiment, the process may be used to deliver a therapeutic polynucleotide to a muscle cell for the treatment of muscular dystrophy. The defective genes that cause MD are known for many forms of the disease. These defective genes either fail to produce a protein product, produce a protein product that fails to function properly, or produce a dysfunctional protein product that interferes with the proper function of the cell. The polynucleotide may encode a therapeutically functional protein or a polynucleotide that inhibits production or activity of a dysfunctional protein. The polynucleotide may be delivered to muscle cells of an MD patient for therapeutic treatment of the disease, for treatment of secondary manifestations of muscular dystrophy, or for other muscular atrophy or injury. Genes that may be expressed from delivered polynucleotides, or inhibited by delivered polynucleotides, may be selected from the list comprising: dystrophin (Duchene's and Becker MD); dystrophin-associated glycoproteins (β-sarcoglycan and δ-sarcoglycan, limb-girdle MD 2E and 2F; α-sarcoglycan and γ-sarcoglycan, limb-girdle MD 2D and 2C), utrophin, calpain (autosomal recessive limb-girdle MD type 2A), caveolin-3 (autosomal-dominant limb-girdle MD), laminin-alpha2 (merosin-deficient congenital MD), fukutin (Fukuyama type congenital MD) and emerin (Emery-Dreifuss MD), myotilin, lamin A/C, calpain-3, dysferlin, telethonin or therapeutic variation of these proteins. A polynucleotide expressing a protein beneficial to a patient suffering from muscular disease or injury or booster genes aimed at alleviating secondary defects of muscle disease may also be delivered to muscle cells of the patient. Such genes may be selected from the list comprising: mini-agrin (to promote basement membrane formation), utrophin, laminin α2, α7 integrins, GalNac transferase, and ADAM12 (to promote cell adhesion and muscle stability), calpastatin (to protect against muscle necrosis), nitric oxide synthase (to ease inflammation), ADAM12 protein (meltrin alpha), IGF-1, dominant negative myostatin and myostatin inhibitors (to promote muscle regeneration and reduce fibrosis), TGF-β (to regulate muscle mass), Nitric Oxide Synthase (to reduce inflammation), actin, titin, muscle creatine kinase, troponin, growth factors (human growth factor and human growth hormone releasing hormone, and vascular endothelial growth factor (VEGF)), insulin, and anti-inflammatory genes. Polynucleotides such as siRNAs and antisense oligonucleotides may also be delivered to create a myostatin blockade or to inhibit myostatin synthesis (to promote muscle growth), inhibit myogenin production (to increase muscle size) or to modify splicing of a defective endogenous gene.
  • The delivered polynucleotide may express a protein or peptide may be secreted or stay within the cell. For proteins and peptides that are secreted, the gene may contain a sequence that codes for a signal peptide. The delivered polynucleotide can also suppress or inhibit expression of an endogenous gene or gene product. Multiple polynucleotides or polynucleotides containing more than one therapeutic gene may be delivered using the described process.
  • Further objects, features, and advantages of the invention will be apparent from the following detailed description when taken in conjunction with the accompanying drawings.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1A-1B. Photomicrographs of muscle sections histochemically stained for β-galactosidase expression. Panel A represents a muscle (pronator teres) with a high level of expression; panel B represents a muscle (abductor pollicis longus) with an average level of expression. Magnification: 160×.
  • FIG. 2A-2C. Expression of β-galactosidase (light grey) and GFP (white) in rat muscle injected intraarterially at different times with the respective expression pDNAs. Panel A (640× magnification) is a low-power field illustrating that expression of β-galactosidase and GFP were typically not co-localized. Panels B and C are high power fields (1600× magnification) that show an example of co-localization (B) and separate expression (C).
  • FIG. 3A-3C. Muscle sections obtained 5 min (A and B) and 1 h (C) after 50 μg of Rh-pDNA in 10 ml of normal saline were injected within 7 sec into the femoral artery of rat without impeding the outflow (A) or impeding outflow (B and C). Arrows indicate Rh-pDNA between cells and arrowheads indicate pDNA inside myofibers. Magnification: 1260×.
  • FIG. 4A-4C. LacZ expression in mouse skeletal muscle seven days following intra-arterial injections of 100 μg pCI-LacZ (A) or pMI-DYS (B and C) in dystrophic mdx mouse (A and B) or normal ICR mouse (C).
  • FIG. 5. Intravascular injection of therapeutic genes into mammalian limbs. Immunohistochemical staining for human dystrophin expression in mdx4cv mouse gastrocnemius muscle (left panel) one week after intravenous injection of 300 μg of a pDNA human dystrophin expression vector in 0.6 ml of NSS (7.5 s injection). Staining in mdx4cv mice injected with pCI-Luc negative control vector is shown in the right panel.
  • FIG. 6A-6B. Illustration of luciferase expression in leg muscles of dystrophic and normal dog after intra-arterial injection of pCI-Luc plasmid under elevated pressure. Panel A shows expression distribution in normal dog. Panel B shows expression distribution in dystrophic dog model.
  • FIG. 7. Paraffin cross sections of the Pronator quadratus muscles stained with hematoxylin and eosin and examined under light microscope. Left panel—Pronator quadratus muscle transfected with VEGF-165 plasmid. Right panel—Pronator quadratus muscle transfected with EPO plasmid. Top left picture (VEGF-165) demonstrates increased number of vessels and interstitial cells (presumably—endothelial cells), as compared to right picture (EPO-control), magnification ×200. Bottom left picture (VEGF-165) demonstrates increased number of vessels, most small arteries and capillaries, as compare to right picture (EPO-control). Arrows indicate obvious vascular structures, magnification ×6300.
  • FIG. 8. Paraffin cross sections of the Pronator quadratus muscles immunostained for endothelial cell marker—CD31, and examined under confocal laser scanning microscope LSM 510, magnification ×400. CD31 marker visualized with Cy3 (black), nuclei with nucleic acid stains To Pro-3. Muscle fibers and red blood cells were visualized by 488 nm laser having autofluorescent emission. Left picture—Pronator quadratus muscle transfected with VEGF-165 plasmid, demonstrates increased of endothelial cells and small vessels, as compare to right picture (EPO-control). The number of CD31 positive cells was increased significantly in VEGF-165 transfected muscle by 61.7% (p<0.001).
  • DETAILED DESCRIPTION
  • We have found that an intravascular route of administration allows a polynucleotide to be delivered to parenchymal cells in a more even distribution than direct parenchymal injections. The efficiency of polynucleotide delivery and expression is increased by increasing the permeability of the tissue's blood vessel. Permeability is increased by one or more of the following: increasing the intravascular pressure, delivering the injection fluid rapidly (injecting the injection fluid rapidly), using a large injection volume, inhibiting vessel fluid flow, and increasing permeability of the vessel wall. Prior to insertion, subsequent to insertion, or concurrent with insertion the permeability of the vessel is increased using an exterior cuff thereby the genetic material is delivered to the parenchymal cell.
  • We describe a process for inserting a polynucleotide into mammalian cells. We have injected a polynucleotide expressing marker genes into limbs of mouse, rat, and dog and caused the polynucleotide to be delivered to and expressed in a large proportion of muscle cells throughout a limb. More particularly we have injected primate limbs and caused the polynucleotide to be delivered and expressed. The expression levels achieved in non-human primates indicate that the procedure is likely to be efficient in humans. It is noteworthy that expression levels in monkeys were similar to expression levels in rats since the efficiency of many prior art gene transfer techniques is less in larger animals. For both the arm and leg injections, blood flow was impeded by a cuff surrounding the arm or leg. The high luciferase and β-galactosidase levels achieved in monkeys indicate that the procedure is likely to be efficient in humans. It is noteworthy that expression levels were similar in monkeys as those levels in rats since the efficiency of many prior art gene transfer techniques is less in larger animals. In addition, we have injected a polynucleotide expressing the dystrophin gene into limbs of both rat and dog MD models and caused the polynucleotide to be expressed in a large proportion of muscle cells throughout a limb.
  • The described process may also be used repetitively in a single mammal. Multiple injections may be used to provide delivery to additional cells, tissues, or muscle groups, to increase delivery to a single tissue, or where multiple treatments are indicated. Multiple injections may be performed in different limbs of the same animal, within the same limb of the animal, within the same vein of the animal, within different veins in the animal (in the same or different limbs). The site of vessel occlusion may also be the same or different for multiple injections in the same animal.
  • The term cuff means a device for impeding fluid flow through mammalian internal blood vessels. However, for purposes of the claims, cuff refers specifically to a device applied exterior to the mammal's skin and touches the skin in a non-invasive manner. In a preferred embodiment, the cuff is a device that applies external pressure to the mammalian skin and thereby pressure is applied internally to the blood vessel walls. The vessel walls, in an area underneath the cuff, are forced to constrict in amount sufficient to impede fluid from flowing at a normal rate. Impeding fluid flow into and out of an area such as a limb, combined with injection of a solution containing polynucleotides, causes vascular pressure and vessel permeability to increase in the area. Thus, the fluid and its contents (including polynucleotides) are urged out of the vessel walls and into the extravascular space. One example of a cuff is a sphygmomanometer which is normally used to measure blood pressure. In a preferred embodiment of this specification, the sphygmomanometer is used to apply pressure to mammalian skin, around a limb, for the purpose of increasing vessel permeability when combined with solution injection into the vessel. Another example is a tourniquet.
  • In yet another preferred embodiment the use of a cuff (or other external pressure device) is combined with the use of a pharmaceutical or biologically-active agent (such as papaverine) to increase vascular permeability.
  • The term intravascular refers to an intravascular route of administration that enables a polymer, oligonucleotide, or polynucleotide to be delivered to cells more evenly distributed than direct injections. Intravascular herein means within an internal tubular structure called a vessel that is connected to a tissue or organ within the body of an animal, including mammals. Within the cavity of the tubular structure, a bodily fluid flows to or from the body part. Examples of bodily fluid include blood, lymphatic fluid, or bile. Examples of vessels include arteries, arterioles, capillaries, venules, sinusoids, veins, lymphatics, and bile ducts. The intravascular route includes delivery through the blood vessels such as an artery or a vein.
  • Afferent blood vessels of organs are defined as vessels in which blood flows toward the organ or tissue under normal physiologic conditions. Efferent blood vessels are defined as vessels in which blood flows away from the organ or tissue under normal physiologic conditions. In the heart, afferent vessels are known as coronary arteries, while efferent vessels are referred to as coronary veins.
  • Proximal is defined herein as a position nearer to the point of attachment or origin or central point, especially located nearer the center of the body, as the proximal part of a limb. For example, the proximal end of the femur is part of the hip joint, the knee is proximal to the toes, the shoulder is proximal to the elbow and the elbow is proximal to the hand. The opposite of proximal is distal.
  • The term naked nucleic acid indicates that the nucleic acids are not associated with a transfection reagent or other delivery vehicle that is required for the nucleic acid to be delivered to a target cell. A transfection reagent is a compound or compounds used in the prior art that mediates nucleic acid's entry into cells.
  • Expression cassette refers to a natural or recombinantly produced nucleic acid which is capable of expressing protein(s). A DNA expression cassette typically includes a promoter (allowing transcription initiation), and a sequence encoding one or more proteins (transgene(s)). Optionally, the expression cassette may include transcriptional enhancers, locus control regions, matrix attachment regions, scaffold attachment regions, non-coding sequences, splicing signals, transcription termination signals, and polyadenylation signals. An RNA expression cassette typically includes a translation initiation codon (allowing translation initiation), and a sequence encoding one or more proteins. Optionally, the expression cassette may include translation termination signals, a polyadenosine sequence, internal ribosome entry sites (IRES), and non-coding sequences.
  • The expression cassette promoter can be selected from any of the known promoters isolated from the group consisting of, but not limited to, the human genome, mammalian genomes, microbial genomes, and chimeric sequences. Additionally, artificially constructed sequences can be used that have shown to have promoter activity in the target cell type. Examples of viral promoters that have successfully been used to express transgenes include: human cytomegalovirus immediate early promoter, Rous sarcoma virus, Moloney leukemia virus, and SV40. Examples of mammalian promoters include: elongation factor 1, muscle creatine kinase, actin, desmin, and troponin. The choice of promoter in conjunction with other expression cassette elements can determine the level of transgene protein production in target cells. The expression cassette can be designed to express preferentially in specific cell types (operationally defined as a 5-fold higher expression level in the specific cell type compared to the average expression level in other cell types). A promoter, or combination of a promoter and other regulatory elements in the expression cassette, resulting in preferential expression in specific cell types is frequently referred to as tissue-specific. An example of a tissue-specific promoter is the muscle creatine kinase promoter, which expresses transgenes at high levels in skeletal muscle cells, whereas expression in other cell types is at lower levels. Preferential expression in muscle cells can be achieved by using promoters and regulatory elements from muscle-specific genes (e.g., muscle creatine kinase, myosin light chain, desmin, skeletal actin), or by combining transcriptional enhancers from muscle-specific genes with a promoter normally active in many cell types (e.g., the human cytomegalovirus immediate early promoter in combination with the myosin light chain enhancer).
  • It may be desirable to regulate expression of the delivered polynucleotide using regulated promoters. Regulated promoters may be inducible or repressible. Regulated gene expression systems may be selected from the list comprising: drug-dependent gene regulation, tetracycline/doxycycline-inducible, tetracycline/doxycycline-repressible, rapamycin-inducible, β-galactoside, streptogramin-regulated, bacterial repressor protein, antiprogestin-inducible GeneSwitch® (Valentis, Inc., induced by mifepristone), nuclear hormone receptor ligand binding domain (antiprogestin-, antiestrogen-, ecdysteroid-, glucocorticoid-responsive), heterodimeric protein, metabolic regulated, hypoxia responsive, and glucose responsive systems. Some of these systems are regulated by proteins naturally occurring in mammalian cells while others require co-delivery of a gene encoding a transcription activator or repressor.
  • It may also be desirable for the delivered polynucleotide to be expressed from a muscle specific promoter. Muscle specific promoters may be selected from the list comprising: muscle creatine kinase (MCK), myosin light chain, myosin light chain 3F, desmin, alpha-actin, enolase, utrophin, dystrophin, sarcoglycan and other dystrophin-associated glycoprotein promoters. Still other transcription elements that function in muscle include: actin and β-actin promoters, E-box elements, MEF-2 elements, TEF-1 elements, SRE sites, myogenin enhancer sequences, and viral promoters such as CMV and SV40.
  • Protein refers herein to a linear series of greater than 2 amino acid residues connected one to another as in a polypeptide. A therapeutic effect of the protein in attenuating or preventing the disease state can be accomplished by the protein either staying within the cell, remaining attached to the cell in the membrane, or being secreted and dissociated from the cell where it can enter the general circulation and blood. Secreted proteins that can be therapeutic include hormones, cytokines, interferons, enzymes (e.g. lysosomal enzymes), growth factors, clotting factors, anti-protease proteins (e.g., alpha1-antitrypsin), angiogenic proteins (e.g., vascular endothelial growth factor, fibroblast growth factors), anti-angiogenic proteins (e.g., endostatin, angiostatin), and other proteins that are present in the blood. Proteins on the membrane can have a therapeutic effect by providing a receptor for the cell to take up a protein or lipoprotein (e.g., low density lipoprotein receptor). Therapeutic proteins that stay within the cell (intracellular proteins) can be enzymes that clear a circulating toxic metabolite as in phenylketonuria. They can also cause a cancer cell to be less proliferative or cancerous (e.g., less metastatic), or interfere with the replication of a virus. Intracellular proteins can be part of the cytoskeleton (e.g., actin, dystrophin, myosins, sarcoglycans, and dystroglycans) and thus have a therapeutic effect in cardiomyopathies and musculoskeletal diseases (e.g., Duchenne muscular dystrophy, limb-girdle disease). Other therapeutic proteins of particular interest to treating heart disease include polypeptides affecting cardiac contractility (e.g., calcium and sodium channels), inhibitors of restenosis (e.g., nitric oxide synthetase), angiogenic factors, and anti-angiogenic factors.
  • Constructs to Improve Secretion of Muscle Expressed Protein into the Blood:
  • Proteins are targeted for secretion from cells by the presence of a signal peptide. During transit through the endoplasmic reticulum, the signal peptide is removed by specific proteolytic cleavage. It can be anticipated that secretion of certain proteins can be improved by replacing the endogenous signal peptide with a heterologous signal peptide. This can be accomplished by exchanging the coding regions for the signal peptides in the nucleic acid. For example, the signal from the protein placental alkaline phosphatase (often used in a truncated from as secreted alkaline phosphatase, SEAP) can be used to replace the signal from the protein factor IX. This may result in better secretion of the factor IX protein from muscle cells. Since the signal peptide is cleaved prior to secretion, the secreted mature factor IX protein is unaltered and functional. Alternatively, one can construct a fusion between the complete SEAP and target protein, or use other defined protein sequence known to enhance transmembrane transport, such as the TAT protein from the human immunodeficiency virus, or the VP22 protein from herpes viruses.
  • There are three types of reporter (or marker) gene products that are expressed from reporter genes. The reporter gene/protein systems include:
  • Intracellular gene products included proteins such as luciferase, β-galactosidase, or chloramphenicol acetyl transferase. Typically, they are enzymes whose enzymatic activity can be easily measured. Intracellular gene products, such as β-galactosidase or green fluorescent protein, can identify cells expressing the reporter gene. On the basis of the intensity of cellular staining, these reporter gene products also yield qualitative information concerning the amount of foreign protein produced per cell. Secreted gene products such as growth hormone, factor IX, secreted alkaline phosphatase, or alpha1-antitrypsin are useful for determining the amount of a secreted protein that a gene transfer procedure can produce. The reporter gene product can be assayed in a small amount of blood.
  • We have disclosed gene expression achieved from reporter genes in specific tissues. The terms therapeutic and therapeutic results are defined in this application as a nucleic acid which is transfected into a cell, in vivo, resulting in a gene product (e.g. protein) being expressed in the cell or secreted from the cell. Levels of a gene product, including reporter (marker) gene products, are measured which then indicate a reasonable expectation of similar amounts of gene expression by transfecting other nucleic acids. Levels of treatment considered beneficial by a person having ordinary skill in the art of gene therapy differ from disease to disease, for example: Hemophilia A and B are caused by deficiencies of the X-linked clotting factors VIII and IX, respectively. Their clinical course is greatly influenced by the percentage of normal serum levels of factor VIII or IX: <2%, severe; 2-5%, moderate; and 5-30% mild. This indicates that in severe patients an increase from 1% to 2% of the normal level can be considered beneficial. Levels greater than 6% prevent spontaneous bleeds but not those secondary to surgery or injury. A person having ordinary skill in the art of gene therapy would reasonably anticipate beneficial levels of expression of a gene specific for a disease based upon sufficient levels of marker gene results. In the hemophilia example, if marker genes were expressed to yield a protein at a level comparable in volume to 2% of the normal level of factor VIII, it can be reasonably expected that the gene coding for factor VIII would also be expressed at similar levels.
  • Parenchymal Cells
  • Parenchymal cells are the distinguishing cells of a gland or organ contained in and supported by the connective tissue framework. The parenchymal cells typically perform a function that is unique to the particular organ. The term parenchymal often excludes cells that are common to many organs and tissues such as fibroblasts and endothelial cells within blood vessels.
  • In a liver organ, the parenchymal cells include hepatocytes, Kupffer cells and the epithelial cells that line the biliary tract and bile ductules. The major constituent of the liver parenchyma are polyhedral hepatocytes (also known as hepatic cells) that presents at least one side to an hepatic sinusoid and opposed sides to a bile canaliculus. Liver cells that are not parenchymal cells include cells within the blood vessels such as the endothelial cells or fibroblast cells. In one preferred embodiment hepatocytes are targeted by injecting the polynucleotide within the tail vein of a rodent such as a mouse.
  • In striated muscle, the parenchymal cells include myoblasts, satellite cells, myotubules, and myofibers. In cardiac muscle, the parenchymal cells include the myocardium also known as cardiac muscle fibers or cardiac muscle cells and the cells of the impulse connecting system such as those that constitute the sinoatrial node, atrioventricular node, and atrioventricular bundle. In a preferred embodiment skeletal muscle, cardiac muscle, or diaphragm muscle is targeted by injecting the polynucleotide into the blood vessel supplying or draining the tissue.
  • Polymers:
  • A polymer is a molecule built up by repetitive bonding together of smaller units called monomers. In this application the term polymer includes both oligomers which have two to about 80 monomers and polymers having more than 80 monomers. The polymer can be linear, branched network, star, comb, or ladder types of polymer. The polymer can be a homopolymer in which a single monomer is used or can be copolymer in which two or more monomers are used. Types of copolymers include alternating, random, block and graft.
  • One of our several methods of nucleic acid delivery to cells is the use of nucleic acid-polycations complexes. It was shown that cationic proteins like histones and protamines or synthetic polymers like polylysine, polyarginine, polyornithine, DEAE dextran, polybrene, and polyethylenimine are effective intracellular delivery agents while small polycations like spermine are ineffective.
  • A polycation is a polymer containing a net positive charge, for example poly-L-lysine hydrobromide. The polycation can contain monomer units that are charge positive, charge neutral, or charge negative, however, the net charge of the polymer must be positive. A polycation also can mean a non-polymeric molecule that contains two or more positive charges. A polyanion is a polymer containing a net negative charge, for example polyglutamic acid. The polyanion can contain monomer units that are charge negative, charge neutral, or charge positive, however, the net charge on the polymer must be negative. A polyanion can also mean a non-polymeric molecule that contains two or more negative charges. The term polyion includes polycation, polyanion, zwitterionic polymers, and neutral polymers that contain equal amounts of anions and cations. The term zwitterionic refers to the product (salt) of the reaction between an acidic group and a basic group that are part of the same molecule. Salts are ionic compounds that dissociate into cations and anions when dissolved in solution. Salts increase the ionic strength of a solution, and consequently decrease interactions between nucleic acids with other cations.
  • In one embodiment, polycations are mixed with polynucleotides for intravascular delivery to a cell. Polycations provide the advantage of allowing attachment of DNA to the target cell surface. The polymer forms a cross-bridge between the polyanionic nucleic acids and the polyanionic surfaces of the cells. As a result the main mechanism of DNA translocation to the intracellular space might be non-specific adsorptive endocytosis which may be more effective then liquid endocytosis or receptor-mediated endocytosis. Furthermore, polycations are a very convenient linker for attaching specific receptors to DNA and as result, DNA-polycation complexes can be targeted to specific cell types.
  • Additionally, polycations protect DNA in complexes against nuclease degradation. This is important for both extra- and intracellular preservation of DNA. The endocytic step in the intracellular uptake of DNA-polycation complexes is suggested by results in which DNA expression is only obtained by incorporating a mild hypertonic lysis step (either glycerol or DMSO). Gene expression is also enabled or increased by preventing endosome acidification with NH4CI or chloroquine. Polyethylenimine which facilitates gene expression without additional treatments probably disrupts endosomal function itself. Disruption of endosomal function has also been accomplished by linking the polycation to endosomal-disruptive agents such as fusion peptides membrane active compounds, or adenoviruses.
  • Membrane Active Compounds
  • Many biologically active compounds, in particular large and/or charged compounds, are incapable of crossing biological membranes. In order for these compounds to enter cells, the cells must either take them up by endocytosis, into endosomes, or there must be a disruption of the cellular membrane to allow the compound to cross. In the case of endosomal entry, the endosomal membrane must be disrupted to allow for the entrance of the compound in the interior of the cell. Therefore, either entry pathway into the cell requires a disruption of the cellular membrane. There exist compounds termed membrane active compounds that disrupt membranes. One can imagine that if the membrane active agent were operative in a certain time and place it would facilitate the transport of the biologically active compound across the biological membrane. The control of when and where the membrane active compound is active is crucial to effective transport. If the membrane active compound is too active or active at the wrong time, then no transport occurs or transport is associated with cell rupture and thereby cell death. Nature has evolved various strategies to allow for membrane transport of biologically active compounds including membrane fusion and the use membrane active compounds whose activity is modulated such that activity assists transport without toxicity. Many lipid-based transport formulations rely on membrane fusion and some membrane active peptides' activities are modulated by pH. In particular, viral coat proteins are often pH-sensitive, inactive at neutral or basic pH and active under the acidic conditions found in the endosome.
  • Polycations also cause DNA condensation. The volume which one DNA molecule occupies in complex with polycations is drastically lower than the volume of a free DNA molecule. The size of DNA/polymer complex may be important for gene delivery in vivo. In terms of intravenous injection, DNA must cross the endothelial barrier and reach the parenchymal cells of interest.
  • The average diameter of liver fenestrae (holes in the endothelial barrier) is about 100 nm, and increases in pressure and/or permeability can increase the size of the fenestrae. The fenestrae size in other organs is usually less. The size of the DNA complexes is also important for the cellular uptake process. After binding to the target cells the DNA-polycation complex is expected to be taken up by endocytosis.
  • Polymers may incorporate compounds that increase their utility. These groups can be incorporated into monomers prior to polymer formation or attached to the polymer after its formation. The gene transfer enhancing signal (Signal) is defined in this specification as a molecule that modifies the nucleic acid complex and can direct it to a cell location (such as tissue cells) or location in a cell (such as the nucleus) either in culture or in a whole organism. By modifying the cellular or tissue location of the foreign gene, the expression of the foreign gene can be enhanced.
  • The gene transfer enhancing signal can be a protein, peptide, lipid, steroid, sugar, carbohydrate, nucleic acid or synthetic compound. The gene transfer enhancing signals enhance cellular binding to receptors, cytoplasmic transport to the nucleus and nuclear entry or release from endosomes or other intracellular vesicles.
  • Nuclear localizing signals enhance the targeting of the gene into proximity of the nucleus and/or its entry into the nucleus. Such nuclear transport signals can be a protein or a peptide such as the SV40 large T ag NLS or the nucleoplasmin NLS. These nuclear localizing signals interact with a variety of nuclear transport factors such as the NLS receptor (karyopherin alpha) which then interacts with karyopherin beta. The nuclear transport proteins themselves could also function as NLS's since they are targeted to the nuclear pore and nucleus.
  • Signals that enhance release from intracellular compartments (releasing signals) can cause DNA release from intracellular compartments such as endosomes (early and late), lysosomes, phagosomes, vesicle, endoplasmic reticulum, Golgi apparatus, trans Golgi network (TGN), and sarcoplasmic reticulum. Release includes movement out of an intracellular compartment into cytoplasm or into an organelle such as the nucleus. Releasing signals include chemicals such as chloroquine, bafilomycin or Brefeldin A1 and the ER-retaining signal (KDEL sequence), viral components such as influenza virus hemagglutinin subunit HA-2 peptides and other types of amphipathic peptides.
  • Cellular receptor signals are any signal that enhances the association of the gene with a cell. This can be accomplished by either increasing the binding of the gene to the cell surface and/or its association with an intracellular compartment, for example: ligands that enhance endocytosis by enhancing binding the cell surface. This includes agents that target to the asialoglycoprotein receptor by using asialoglycoproteins or galactose residues. Other proteins such as insulin, EGF, or transferrin can be used for targeting. Peptides that include the RGD sequence can be used to target many cells. Chemical groups that react with sulfhydryl or disulfide groups on cells can also be used to target many types of cells. Folate and other vitamins can also be used for targeting. Other targeting groups include molecules that interact with membranes such as lipids fatty acids, cholesterol, dansyl compounds, and amphotericin derivatives. In addition viral proteins could be used to bind cells.
  • Polynucleotides
  • The term polynucleotide, or nucleic acid, is a term of art that refers to a polymer containing at least two nucleotides. Nucleotides are the monomeric units of polynucleotide polymers. Polynucleotides with less than 120 monomeric units are often called oligonucleotides. Natural nucleic acids have a deoxyribose- or ribose-phosphate backbone. An artificial or synthetic polynucleotide is any polynucleotide that is polymerized in vitro or in a cell free system and contains the same or similar bases but may contain a backbone of a type other than the natural ribose-phosphate backbone. These backbones include: PNAs (peptide nucleic acids), phosphorothioates, phosphorodiamidates, morpholinos, and other variants of the phosphate backbone of native nucleic acids. Bases include purines and pyrimidines, which further include the natural compounds adenine, thymine, guanine, cytosine, uracil, inosine, and natural analogs. Synthetic derivatives of purines and pyrimidines include, but are not limited to, modifications which place new reactive groups such as, but not limited to, amines, alcohols, thiols, carboxylates, and alkylhalides. The term base encompasses any of the known base analogs of DNA and RNA including, but not limited to, 4-acetylcytosine, 8-hydroxy-N6-methyladenosine, aziridinylcytosine, pseudoisocytosine, 5-(carboxyhydroxylmethyl)uracil, 5-fluorouracil, 5-bromouracil, 5-carboxymethylaminomethyl-2-thiouracil, 5-carboxy-methylaminomethyluracil, dihydrouracil, inosine, N6-isopentenyladenine, 1-methyladenine, 1-methylpseudo-uracil, 1-methylguanine, 1-methylinosine, 2,2-dimethyl-guanine, 2-methyladenine, 2-methylguanine, 3-methyl-cytosine, 5-methylcytosine, N6-methyladenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxy-amino-methyl-2-thiouracil, β-D-mannosylqueosine, 5′-methoxycarbonylmethyluracil, 5-methoxyuracil, 2-methyl-thio-N6-isopentenyladenine, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid, oxybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, N-uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid, pseudouracil, queosine, 2-thiocytosine, and 2,6-diaminopurine. The term polynucleotide includes deoxyribonucleic acid (DNA) and ribonucleic acid (RNA) and combinations on DNA, RNA and other natural and synthetic nucleotides.
  • DNA may be in form of cDNA, in vitro polymerized DNA, plasmid DNA, parts of a plasmid DNA, genetic material derived from a virus, linear DNA, vectors (P1, PAC, BAC, YAC, and artificial chromosomes), expression cassettes, chimeric sequences, recombinant DNA, chromosomal DNA, an oligonucleotide, anti-sense DNA, or derivatives of these groups. RNA may be in the form of oligonucleotide RNA, tRNA (transfer RNA), snRNA (small nuclear RNA), rRNA (ribosomal RNA), mRNA (messenger RNA), in vitro polymerized RNA, recombinant RNA, chimeric sequences, anti-sense RNA, siRNA (small interfering RNA), ribozymes, or derivatives of these groups. An anti-sense polynucleotide is a polynucleotide that interferes with the function of DNA and/or RNA. Antisense polynucleotides include, but are not limited to: morpholinos, 2′-O-methyl polynucleotides, DNA, RNA and the like. SiRNA comprises a double stranded structure typically containing 15-50 base pairs and preferably 21-25 base pairs and having a nucleotide sequence identical or nearly identical to an expressed target gene or RNA within the cell. Interference may result in suppression of expression. The polynucleotide can be a sequence whose presence or expression in a cell alters the expression or function of cellular genes or RNA. In addition, DNA and RNA may be single, double, triple, or quadruple stranded. Double, triple, and quadruple stranded polynucleotide may contain both RNA and DNA or other combinations of natural and/or synthetic nucleic acids.
  • A polynucleotide can be delivered to a cell to express an exogenous nucleotide sequence, to inhibit, eliminate, augment, or alter expression of an endogenous nucleotide sequence, or to express a specific physiological characteristic not naturally associated with the cell. Polynucleotides may be coded to express a whole or partial protein, or may be anti-sense.
  • A delivered polynucleotide can stay within the cytoplasm or nucleus apart from the endogenous genetic material. Alternatively, the polymer could recombine (become a part of) the endogenous genetic material. For example, DNA can insert into chromosomal DNA by either homologous or non-homologous recombination.
  • RNA function inhibitor. A RNA function inhibitor comprises any nucleic acid or nucleic acid analog containing a sequence whose presence or expression in a cell causes the degradation of or inhibits the function or translation of a specific cellular RNA, usually a mRNA, in a sequence-specific manner. An RNA function inhibitor may also inhibit the transcription of a gene into RNA. Inhibition of RNA can thus effectively inhibit expression of a gene from which the RNA is transcribed. RNA function inhibitors are selected from the group comprising: siRNA, interfering RNA or RNAi, dsRNA, RNA Polymerase III transcribed DNAs, ribozymes, and antisense nucleic acid, which may be RNA, DNA, or artificial nucleic acid. SiRNA comprises a double stranded structure typically containing 15-50 base pairs and preferably 21-25 base pairs and having a nucleotide sequence identical or nearly identical to an expressed target gene or RNA within the cell. Antisense polynucleotides include, but are not limited to: morpholinos, 2′-O-methyl polynucleotides, DNA, RNA and the like. RNA polymerase III transcribed DNAs contain promoters, such as the U6 promoter. These DNAs can be transcribed to produce small hairpin RNAs in the cell that can function as siRNA or linear RNAs that can function as antisense RNA. The RNA function inhibitor may be polymerized in vitro, recombinant RNA, contain chimeric sequences, or derivatives of these groups. The RNA function inhibitor may contain ribonucleotides, deoxyribonucleotides, synthetic nucleotides, or any suitable combination such that the target RNA and/or gene is inhibited. In addition, these forms of nucleic acid may be single, double, triple, or quadruple stranded.
  • Vectors are polynucleic molecules originating from a virus, a plasmid, or the cell of a higher organism into which another nucleic fragment of appropriate size can be integrated; vectors typically introduce foreign DNA into host cells, where it can be reproduced. Examples are plasmids, cosmids, and yeast artificial chromosomes; vectors are often recombinant molecules containing DNA sequences from several sources. A vector includes a viral vector: for example, adenovirus; DNA; adenoassociated viral vectors (AAV) which are derived from adenoassociated viruses and are smaller than adenoviruses; and retrovirus (any virus in the family Retroviridae that has RNA as its nucleic acid and uses the enzyme reverse transcriptase to copy its genome into the DNA of the host cell's chromosome; examples include VSV G and retroviruses that contain components of lentivirus including HIV type viruses).
  • A vector is used in this specification to mean any DNA molecule which could include associate molecules to transfer DNA sequences into a cell for expression. Examples include naked DNA, non-viral DNA complexes (e.g. DNA plus polymers (cationic or anionic), DNA plus transfection enhancing compounds, and DNA plus amphipathic compounds) and viral particles.
  • A non-viral vector is defined as a vector that is not assembled within a eukaryotic cell including non-viral DNA/polymer complexes, DNA with transfection enhancing compounds and DNA+amphipathic compounds.
  • Skin is the external covering of a mammalian body including the epidermis, the dermis, and the subcutaneous tissue.
  • Permeability
  • In another preferred embodiment, the permeability of the vessel is increased. Efficiency of polynucleotide delivery and expression was increased by increasing the permeability of a blood vessel within the target tissue. Permeability is defined here as the propensity for macromolecules, such as polynucleotides, to move through vessel walls and enter the extravascular space. One measure of permeability is the rate at which macromolecules move through the vessel wall and out of the vessel. Another measure of permeability is the lack of force that resists the movement of polynucleotides being delivered to leave the intravascular space.
  • To obstruct, in this specification, is to block or inhibit inflow or outflow of blood in a vessel. Rapid injection may be combined with obstructing the outflow to increase permeability. For example, an afferent vessel supplying an organ is rapidly injected and the efferent vessel draining the tissue is ligated transiently. The efferent vessel (also called the venous outflow or tract) draining outflow from the tissue is also partially or totally impeded for a period of time sufficient to allow delivery of a polynucleotide. In the reverse, an efferent is injected and an afferent vessel flow is impeded.
  • In another preferred embodiment, the intravascular pressure of a blood vessel is increased by increasing the osmotic pressure within the blood vessel. Typically, hypertonic solutions containing salts such as NaCl, sugars or polyols such as mannitol are used. Hypertonic means that the osmolarity of the injection solution is greater than physiologic osmolarity. Isotonic means that the osmolarity of the injection solution is the same as the physiological osmolarity (the tonicity or osmotic pressure of the solution is similar to that of blood). Hypertonic solutions have increased tonicity and osmotic pressure similar to the osmotic pressure of blood and cause cells to shrink.
  • In another preferred embodiment, the permeability of the blood vessel can also be increased by a biologically-active molecule. A biologically-active molecule is a protein or a simple chemical such as papaverine or histamine that increases the permeability of the vessel by causing a change in function, activity, or shape of cells within the vessel wall such as the endothelial or smooth muscle cells. Typically, biologically-active molecules interact with a specific receptor or enzyme or protein within the vascular cell to change the vessel's permeability. Biologically-active molecules include vascular permeability factor (VPF) which is also known as vascular endothelial growth factor (VEGF). Another type of biologically-active molecule can also increase permeability by changing the extracellular connective material. For example, an enzyme could digest the extracellular material and increase the number and size of the holes of the connective material. Another type of biologically-active molecule is a chelator that binds calcium and thereby increases the endothelium permeability.
  • In another embodiment a non-viral vector along with a polynucleotide is intravascularly injected in a large injection volume. The injection volume is dependent on the size of the animal to be injected and can be from 1.0 to 3.0 ml or greater for small animals (i.e. tail vein injections into mice). The injection volume for rats can be from 6 to 35 ml or greater. The injection volume for primates can be 70 to 200 ml or greater. The injection volumes in terms of ml/body weight can be 0.03 ml/g to 0.1 ml/g or greater.
  • The injection volume can also be related to the target tissue. For example, delivery of a non-viral vector with a polynucleotide to a limb can be aided by injecting a volume greater than 5 ml per rat limb or greater than 70 ml for a primate. The injection volumes in terms of ml/limb muscle are usually within the range of 0.6 to 1.8 ml/g of muscle but can be greater. In another example, delivery of a polynucleotide to liver in mice can be aided by injecting the non-viral vector-polynucleotide in an injection volume from 0.6 to 1.8 ml/g of liver or greater. In another preferred embodiment, delivering a polynucleotide-non-viral vector to a limb of a primate rhesus monkey, the complex can be in an injection volume from 0.6 to 1.8 ml/g of limb muscle or anywhere within this range.
  • In another embodiment the injection fluid is injected into a vessel rapidly. The speed of the injection is partially dependent on the volume to be injected, the size of the vessel to be injected into, and the size of the animal. In one embodiment the total injection volume (1-3 mls) can be injected from 5 to 15 seconds into the vascular system of mice. In another embodiment the total injection volume (6-35 mls) can be injected into the vascular system of rats from 20 to 7 seconds. In another embodiment the total injection volume (80-200 mls) can be injected into the vascular system of monkeys from 120 seconds or less.
  • In another embodiment a large injection volume is used and the rate of injection is varied. Injection rates of less than 0.012 ml per gram (animal weight) per second are used in this embodiment. In another embodiment injection rates of less than ml per gram (target tissue weight) per second are used for gene delivery to target organs. In another embodiment injection rates of less than 0.06 ml per gram (target tissue weight) per second are used for gene delivery into limb muscle and other muscles of primates.
  • Cleavable Polymers
  • A prerequisite for gene expression is that once DNA/polymer complexes have entered a cell the polynucleotide must be able to dissociate from the cationic polymer. This may occur within cytoplasmic vesicles (i.e. endosomes), in the cytoplasm, or the nucleus. We have developed bulk polymers prepared from disulfide bond containing co-monomers and cationic co-monomers to better facilitate this process. These polymers have been shown to condense polynucleotides, and to release the nucleotides after reduction of the disulfide bond. These polymers can be used to effectively complex with DNA and can also protect DNA from DNases during intravascular delivery to the liver and other organs. After internalization into the cells the polymers are reduced to monomers, effectively releasing the DNA, as a result of the stronger reducing conditions (glutathione) found in the cell. Negatively charged polymers can be fashioned in a similar manner, allowing the condensed nucleic acid particle (DNA plus polycation) to be recharged with a cleavable anionic polymer resulting in a particle with a net negative charge that after reduction of disulfide bonds will release the polynucleic acid. The reduction potential of the disulfide bond in the reducible co-monomer can be adjusted by chemically altering the disulfide bonds environment. This will allow the construction of particles whose release characteristics can be tailored so that the polynucleic acid is released at the proper point in the delivery process.
  • pH Cleavable Polymers for Intracellular Compartment Release
  • A cellular transport step that has importance for gene transfer and drug delivery is that of release from intracellular compartments such as endosomes (early and late), lysosomes, phagosomes, vesicle, endoplasmic reticulum, Golgi apparatus, trans Golgi network (TGN), and sarcoplasmic reticulum. Release includes movement out of an intracellular compartment into cytoplasm or into an organelle such as the nucleus. Chemicals such as chloroquine, bafilomycin or Brefeldin A1 may increase release of molecules into the cytoplasm of a cell. Chloroquine decreases the acidification of the endosomal and lysosomal compartments but also affects other cellular functions. Brefeldin A, an isoprenoid fungal metabolite, collapses reversibly the Golgi apparatus into the endoplasmic reticulum and the early endosomal compartment into the trans-Golgi network (TGN) to form tubules. Bafilomycin A1, a macrolide antibiotic is a more specific inhibitor of endosomal acidification and vacuolar type H+-ATPase than chloroquine. The ER-retaining signal (KDEL sequence) has been proposed to enhance delivery to the endoplasmic reticulum and prevent delivery to lysosomes.
  • To increase the stability of DNA particles in serum, we have added to positively-charged DNA-polycation particles polyanions that form a third layer in the DNA complex and make the particle negatively charged. To assist in the disruption of the DNA complexes, we have synthesized polymers that are cleaved in the acid conditions found in the endosome, pH 5-7. We also have reason to believe that cleavage of polymers in the DNA complexes in the endosome assists in endosome disruption and release of DNA into the cytoplasm.
  • There are two ways to cleave a polyion: cleavage of the polymer backbone resulting in smaller polyions or cleavage of the link between the polymer backbone and the ion containing groups resulting in small ionized molecules and a polymer. In either case, the interaction between the polyion and DNA is broken and the number of molecules in the endosome increases. This causes an osmotic shock to the endosomes and disrupts the endosomes. In the second case, if the polymer backbone is hydrophobic it may interact with the membrane of the endosome. Either effect may disrupt the endosome and thereby assist in release of DNA.
  • To construct cleavable polymers, one may attach the ions or polyions together with bonds that are inherently labile such as disulfide bonds, diols, diazo bonds, ester bonds, sulfone bonds, acetals, ketals, enol ethers, enol esters, imines, imminiums, and enamines Another approach is construct the polymer in such a way as to put reactive groups, i.e. electrophiles and nucleophiles, in close proximity so that reaction between the function groups is rapid. Examples include having carboxylic acid derivatives (acids, esters, amides) and alcohols, thiols, carboxylic acids or amines in the same molecule reacting together to make esters, thiol esters, acid anhydrides or amides.
  • The present invention additionally provides for the use of polymers containing silicon-nitrogen (silazanes) linkages (either in the main chain of the polymer or in a side chain of the polymer) that are susceptible to hydrolysis. Hydrolysis of a silazane leads to the formation of a silanol and an amine. Silazanes are inherently more susceptible to hydrolysis than is the silicon-oxygen-carbon linkage; however, the rate of hydrolysis is increased under acidic conditions. The substitution on both the silicon atom and the amine can affect the rate of hydrolysis due to steric and electronic effects. This allows for the possibility of tuning the rate of hydrolysis of the silizane by changing the substitution on either the silicon or the amine to facilitate the desired affect.
  • In one embodiment, ester acids and amide acids that are labile in acidic environments (pH less than 7, greater than 4) to form an alcohol and amine and an anhydride are use in a variety of molecules and polymers that include peptides, lipids, and multimolecular associations such as liposomes.
  • In one embodiment, ketals that are labile in acidic environments (pH less than 7, greater than 4) to form a diol and a ketone are use in a variety of molecules and polymers that include peptides, lipids, and liposomes.
  • In one embodiment, acetals that are labile in acidic environments (pH less than 7, greater than 4) to form a diol and an aldehyde are use in a variety of molecules and polymers that include peptides, lipids, and liposomes.
  • In one embodiment, enols that are labile in acidic environments (pH less than 7, greater than 4) to form a ketone and an alcohol are use in a variety of molecules and polymers that include peptides, lipids, and liposomes.
  • In one embodiment, iminiums that are labile in acidic environments (pH less than 7, greater than 4) to form an amine and an aldehyde or a ketone are use in a variety of molecules and polymers that include peptides, lipids, and liposomes.
  • pH-Sensitive Cleavage of Peptides and Polypeptides:
  • In one embodiment, peptides and polypeptides (both referred to as peptides) are modified by an anhydride. The amine (lysine), alcohol (serine, threonine, tyrosine), and thiol (cysteine) groups of the peptides are modified by an anhydride to produce an amide, ester or thioester acid. In the acidic environment of the internal vesicles (pH less than 6.5, greater than 4.5) (early endosomes, late endosomes, or lysosome) the amide, ester, or thioester is cleaved displaying the original amine, alcohol, or thiol group and the anhydride.
  • A variety of endosomolytic and amphipathic peptides can be used in this embodiment. A positively-charged amphipathic/endosomolytic peptide is converted to a negatively-charged peptide by reaction with the anhydrides to form the amide acids and this compound is then complexed with a polycation-condensed nucleic acid. After entry into the endosomes, the amide acid is cleaved and the peptide becomes positively charged and is no longer complexed with the polycation-condensed nucleic acid and becomes amphipathic and endosomolytic. In one embodiment the peptides contains tyrosines and lysines. In yet another embodiment, the hydrophobic part of the peptide (after cleavage of the ester acid) is at one end of the peptide and the hydrophilic part (e.g. negatively charged after cleavage) is at another end. The hydrophobic part could be modified with a dimethylmaleic anhydride and the hydrophilic part could be modified with a citranconyl anhydride. Since the dimethylmaleyl group is cleaved more rapidly than the citraconyl group, the hydrophobic part forms first. In another embodiment the hydrophilic part forms alpha helixes or coil-coil structures.
  • pH-Sensitive Cleavage of Lipids and Liposomes
  • In another embodiment, the ester, amide or thioester acid is complexed with lipids and liposomes so that in acidic environments the lipids are modified and the liposome becomes disrupted, fusogenic or endosomolytic. The lipid diacylglycerol is reacted with an anhydride to form an ester acid. After acidification in an intracellular vesicle the diacylglycerol reforms and is very lipid bilayer disruptive and fusogenic.
  • Non-Cleavable Polymers
  • Many cationic polymers such as histone (H1, H2a, H2b, H3, H4, H5), HMG proteins, poly-L-lysine, polyethylenimine, protamine, and poly-histidine are used to compact polynucleic acids to help facilitate gene delivery in vitro and in vivo. A key for efficient gene delivery using prior art methods is that the non-cleavable cationic polymers (both in vitro and in vivo) must be present in a charge excess over the DNA so that the overall net charge of the DNA/polycation complex is positive. Conversely, using our tail vein injection process having non-cleavable cationic polymer/DNA complexes we found that gene expression is most efficient when the overall net charge of the complexes are negative (DNA negative charge>polycation positive charge). Tail vein injections using cationic polymers commonly used for DNA condensation and in vitro gene delivery revealed that high gene expression occurred when the net charge of the complexes were negative.
  • Angiogenesis
  • The term, angiogenesis, in this specification is defined as any formation of new blood vessels. Angiogenesis may also refer to the sprouting of new blood vessels (endothelium-lined channels such as capillaries) from pre-existing vessels as a result of proliferation and migration of endothelial cells. The maturation or enlargement of vessels via recruitment of smooth muscle cells, i.e. the formation of collateral arteries from pre-existing arterioles, is termed arteriogenesis. Vasculogenesis refers to the in situ formation of blood vessels from angioblasts and endothelial precursor cells (EPCs). An anastomosis is a connection between two blood vessels. The formation of anastomoses can be important for restoring blood flow to ischemic tissue. The formation of new vessels in ischemic tissue or in other tissue with insufficient blood perfusion is termed revascularization. As used herein, the term angiogenesis encompasses arteriogenesis, vasculogenesis, anastomosis formation, and revascularization.
  • Angiogenesis is regulated by soluble secreted factors, cell surface receptors and transcription factors. Secreted factors include cytokines, chemokines, and growth factors that affect endothelial cells, smooth muscle cells, monocytes, leukocytes, and precursor cells. Such factors include: vascular endothelial growth factors, fibroblast growth factors, hepatocyte growth factors, angiopoietin 1 (Ang-1), angiopoietin 2 (Ang-2), Platelet derived growth factors (PDFGs), granulocyte macrophage-colony stimulating factor, insulin-like growth factor-1 (IGF-1), IGF-2, early growth response factor-1 (EGR-1), and human tissue kallikrein (HK).
  • Delivery of genes that encode angiogenic factors to cells in vivo provides an attractive alternative to repetitive injections of protein for the treatment of vascular insufficiency or occlusions. Genes that encode angiogenic factors, including both natural and recombinant secreted factors, receptors, and transcription factors, can be targeted to cells in the affected area, thereby limiting deleterious effects associated with delivering angiogenic factors throughout the body. In particular, according to the described invention, genes for angiogenic factors can be delivered to muscle cells in vivo, including skeletal and cardiac muscle cells. Expression of the gene and secretion of the gene product then induces angiogenesis and improves collateral blood flow in the targeted tissue. The improved blood flow can both improve muscle tissue function and relieve pain associated with vascular diseases.
  • EXAMPLES
  • The high luciferase and β-galactosidase levels achieved in monkeys indicate that the procedure is likely to be efficient in humans. Expression levels were somewhat higher in monkeys than in rats.
  • The intra-arterial procedure requires that blood flow be impeded for substantially less than the couple of hours of ischemia required for tissue damage. In fact, a common anesthesia for human limb surgery (e.g., carpal tunnel repair) involves the blockage of blood flow for over one hour. We have not observed any widespread histologic evidence of ischemic muscle damage in rats or primates following the injections. The minimal elevations of muscle-derived enzymes in the serum also argues against any consequential muscle damage.
  • Given that ˜150 ml of fluid is administered to ˜10 kg animals, the large amount of fluid could adversely affect the animal's cardiovascular or hemodynamic status. However, no adverse effects on the animals were observed.
  • The intravascular pressure can be damaging to the arteries. We have observed minimal intimal changes in the arteries that are presumed to be transient and without consequence. Nonetheless, this minimal arterial damage may be prevented by better controlling the intravascular pressure.
  • For this pDNA administration procedure, several factors limit expression to the non-target tissue. 1) The tourniquet prevents the immediate spread of vector outside of the limb. 2) Efficient pDNA expression in the non-vascular parenchymal cells requires extravasation of the injected pDNA.
  • The procedure requires relatively large amounts of pDNA to be administered. This has not been associated with any toxic effects in rodents or monkeys. Given that the tourniquet delays pDNA distribution outside of the limb and the intravascular pDNA is rapidly degraded by circulating DNases, pDNA toxicity is unlikely. In addition, the cost for producing clinical grade pDNA is considerably less expensive than viral vectors and does not represent an obstacle to its clinical use.
  • Example 1
  • Intraarterial Injections in Monkeys. Seven Rhesus macaque monkeys (5 males; 2 females) of 6 to 13.7 kg body weight underwent intraarterial injections in their limbs following anesthesia with ketamine and halothane. For the forearm injections, a longitudinal incision, ˜3 cm in length, was made on the skin along the inside edge of the biceps brachii and 2 cm above the elbow. After separating the artery from surrounding tissues and veins, a 20 g catheter was inserted into the brachial artery anterogradely and ligated in place. For the lower leg injections, the procedure was essentially the same as that used in the arm, but the incision was located on the upper edge of the popliteal fossae and the 20 g catheter was inserted into the popliteal artery.
  • For both the arm and leg injections, blood flow was impeded by a sphygmomanometer cuff surrounding the arm or leg proximal to the injection site. After the sphygmomanometer was inflated to more than 300 mmHg air pressure, the catheterized vessels were injected with 30 ml of normal saline containing 5 mg papaverine (Sigma Co.). Five min. later, a saline solution containing 100 μg pDNA/ml solution was rapidly injected within 30 to 45 sec. For the arms, the volume of each injection was 75 ml and 90 ml in the first two animals and 120 ml thereafter. The injection volume was 180 ml for the lower legs. The DNA solutions were injected using a nitrogen-pressurized cylinder. Two min after injection, the catheters were removed and the sphygmomanometer deflated.
  • The procedure was initially done on four monkeys in which one arm and leg was injected and muscle biopsies were taken at one (#1-3) or two weeks (#4). Monkey #2 had to be sacrificed at two weeks after injection because of an eye infection (unrelated to our procedure). Three more monkeys (#5-7) received an injection in all four extremities (one arm and leg on one day and the other two extremities two days later). Muscle biopsies were obtained at one week and the animals were sacrificed at two weeks after the injections. In monkeys #6 and #7, an arm and leg were injected with pCI-LacZ; all other injections were with pCI-Luc+.
  • Example 2
  • Reporter Gene Systems. The pCI-Luc+ (Promega, Madison, Wis.) and pCI-LacZ plasmids express a cytoplasmic luciferase and the Escherichia coli LacZ, respectively, from the human cytomegalovirus (CMV) immediate-early promoter. The pCI vector (Promega) also contains an SV40 polyadenylation signal. pMI-Luc+ was constructed by replacing the CMV promoter in pCI-Luc+ with a 3300-bp murine muscle creatine kinase promoter. The vector pEBFP-N1 expresses a nuclear-localizing, blue-shifted green fluorescent protein (GFP) from the CMV promoter (Clontech, Palo Alto, Calif.).
  • Luciferase assays were performed on muscle biopsies, entire muscles and various tissues as previously reported. The relative light units (RLU) were converted to nanograms of luciferase by using luciferase standards (Molecular Probes, Eugene, Oreg.) and a standard curve in which luciferase protein (pg)=RLU×5.1×10−5.
  • For the β-galactosidase assays, muscle samples were taken from the proximal, middle, and distal positions of each muscle, cut into small pieces, frozen in cold isopentane, and stored at −80° C. Muscle pieces were randomly chosen from each muscle sample (for every position) and 10 μm-thick cryostat sections were made. Every tenth section, for a total of 20 sections, was stained and analyzed. The sections were incubated in X-gal staining solution (5 mM potassium ferricyanide, 5 mM potassium ferrocyanide, 1 mM magnesium chloride, 1 mM X-gal in 0.1 M PBS, pH 7.6) for 4-8 hours at room temperature and counterstained with hematoxylin and eosin. Three sections were selected randomly from the 20 sections of each position (usually the 4th, 11th and 17th sections, but an adjacent section was used if these sections were not intact). As previously described, the number of β-galactosidase-positive and total cells were determined within a cross area in each section by moving the counter grid from the top edge of the section to the bottom and from the left edge to the right. The percentage of β-galactosidase-positive cells for each muscle was gotten from the result of positive number divided by total cell number. A weighted average for the percent of transfected cells for each extremity muscle was determined as follows: (ΣAi*Mi)/M where Ai is percent of transfected cells for one muscle, Mi—weight of that muscle and M—whole weight of all muscles.
  • For the co-localization of β-galactosidase and GFP expression, 10 μm-thick cryostat sections were fixed with 4% formaldehyde for 5-10 min. Mouse-anti-β-galactosidase antibody and TRITC-labeled goat-anti-mouse IgG (Sigma) were used as primary and secondary antibodies, respectively. Using a Nikon Optiphot epifluorescence microscope with a SenSys CCD Camera (Photometrics, Tucson, Ariz.), two images were collected from the same view for TRITC-labeled β-galactosidase and for GFP and merged together using the program Adobe Photoshop 4.0.
  • Example 3
  • Luciferase and β-galactosidase expression. Seven rhesus macaque (6-20 years old) received pDNA injections into their limb arteries. All seven monkeys tolerated the procedure well and had full function of their arms, hands, legs and feet following the procedure. In particular, this indicates lack of damage to the radial nerve, which could have been sensitive to the inflated sphygmomanometer surrounding the upper arm. Swelling in the target limbs, a putative correlate of successful gene transfer, was noted afterwards but completely subsided by the next day. When the monkeys awakened from the anesthesia 15 to 30 min after the procedure, they did not appear to be in any discomfort beyond that of normal surgical recovery. Occasionally, the skin in the target limb had some spots of hemorrhage that resolved within several days.
  • Four of the monkeys were sacrificed at 14 to 16 days after injection and all the target muscles of their limbs were assessed for either luciferase or β-galactosidase expression (Table 1). These results indicate that the intra-arterial injection of pCI-Luc+ DNA yielded levels of luciferase expression in all muscles of forearm, hand, lower leg and foot, ranging from 345 to 7332 ng/g muscle (Table 1). The variability in luciferase expression in arm muscles for different animals appears dependent upon whether the tip of the catheter was positioned in the radial or ulnar artery. The average luciferase expression levels in the limb muscles were 991.5±187 ng/g for the arm and 1186±673 ng/g for the leg.
  • After intraarterial injection of pCI-LacZ DNA, β-galactosidase expression was found in myofibers. Large numbers of β-galactosidase-positive myofibers were found in both leg and arm muscles, ranging from less than 1% to more than 30% in different muscles (Table 1 and FIG. 1). The average percentage for all four limbs injected was 7.4%, ranging from 6.3% to 9.9% for each of the limbs. The β-galactosidase percentages for specific muscle groups positively correlated with the luciferase levels in the same muscles (r=0.79).
    TABLE 1
    Mean muscle β-galactosidase or luciferase expression in four muscles from monkeys
    sacrificed two weeks after injection of pCI-LacZ or pCI-Luc+.
    A. Arm muscles
    β-galactosidase (% Luciferase
    positive) (ng/g muscle)
    Muscle group Muscle name (n = 2) (n = 5)
    Anterior Superficial palmaris longus 5.9 ± 0.9 2368 ± 1309
    group group pronator teres 19.9 ± 9.4  1818 ± 336 
    flexor carpi radialis 7.8 ± 0.7 1885 ± 762 
    flexor carpi ulnaris 3.8 ± 3.0 852 ± 314
    flexor digitorum spf. 7.7 ± 1.2 1009 ± 189 
    Deep flexor digitorum prof. 1.0 ± 0.5 544 ± 360
    group pronator quadratus 14.3 ± 11.1 1884 ± 331 
    Posterior Superficial brachioradialis 9.0 ± 8.7 1148 ± 942 
    group group extensor carpi radialis longus 6.6 ± 6.3 1179 ± 584 
    extensor carpi radialis brevis 9.4 ± 4.5 1118 ± 325 
    extensor digitorum 6.2 ± 5.4 1184 ± 94 
    anconeus 2.0 ± 0.3 1744 ± 372 
    extensor carpi ulnaris 0.6 ± 0.4 371 ± 86 
    extensor pollicis longus 6.9 ± 4.3 927 ± 228
    Deep supinator 15.1 ± 9.3  2398 ± 748 
    group abductor pollicis longus 6.2 ± 3.8 927 ± 228
    extensor digiti secund et teriti 6.0 ± 5.5 642 ± 168
    extensor digiti quart et minimi 4.0 ± 3.5 593 ± 140
    Muscles of hand muscle of thumb 15.7 ± 0.5  904 ± 494
    interosseus 17.3 ± 4.3  1974 ± 185 
    Weighted Average  6.3 ± 0.04 991 ± 187
    B. Leg muscles
    β-galactosidase Luciferase
    (%) (ng/g muscle)
    Muscle group Muscle name (n = 2) (n = 2)
    Posterior Superficial gastrocnemius 3.0 ± 2.5 743 ± 33 
    group group soleus 21.2 ± 1.4  2888 ± 2151
    Deep popliteus 37.1 ± 0.5  4423 ± 2657
    group flexor digitorum longus 8.9 ± 2.4 3504 ± 2151
    flexor hallucis longus 9.7 ± 2.4 1355 ± 1224
    tibialis posterior 28.7 ± 4.3  7332 ± 5117
    Anterior group tibialis anterior 2.8 ± 0.2 716 ± 162
    extensor hallucis longus 4.2 ± 1.4 810 ± 497
    extensor digitorum longus 10.9 ± 1.0  3187 ± 1166
    abductor hallucis longus 2.2 ± 0.2 345 ± 104
    Internal group peronaus longus 6.3 ± 2.5 626 ± 383
    peronaus brevis 8.9 ± 1.3 1300 ± 23 
    Muscles of foot extensor digitorum brevis 6.2 ± 5.0 672 ± 607
    extensor hallucis brevis 2.4 ± 1.8 672 ± 607
    LEG MUSCLES Weighted Average 7.3 ± 0.1 1692 ± 768 

    “±” indicates standard error;

    n indicates the number of limbs assayed.
  • Example 4
  • Toxicity. Serum chemistries and histologic analyses were performed to determine if the procedure caused any adverse effects in the monkeys. The serum levels of creatine phosphate kinase (CK), alanine aminotransferase, aspartate aminotransferase (AST) and lactate dehydrogenase (LDH) after surgery were several times higher than before surgery. Levels peaked at 48 hours post-injection and returning to normal within several days. Other serum enzymes such as γ-glutamyltransferase (GGT) and alkaline phosphatase, hematological assays (hematocrit and RBC indices, platelets), serum electrolytes (Na, Cl, K), serum minerals (calcium, phosphate, iron), serum proteins (albumin, total protein), serum lipids (cholesterol, triglycerides), renal indices (urea, creatinine), and bilirubin were unaffected. Total WBC increased within the typical range post-surgery.
  • Limb muscles were obtained 14 to 16 days after intraarterial injection and examined histologically. The vast majority of muscle tissue was well preserved and did not show any sign of pathology. In a few sections, mononuclear cells were noted surrounding β-galactosidase positive myofibers, some of which were undergoing degeneration. Immunostaining for CD-markers indicated that the majority of infiltrating cells were CD3-positive (T lymphocytes) with only a few B cells.
  • Example 5
  • Time course of Muscle Expression After Intravascular Injection in Rats. Muscle luciferase expression was measured at several time points following intravascular delivery of the luciferase gene under control of either the CMV promoter (pCI-Luc+) or MCK promoter (pMI-Luc+) into: a) untreated rats, b) rats continuously immunosuppressed (treated with 2.5 mg/kg of FK506 orally and 1 mg/kg dexamethasone subcutaneously one day prior to, one hour prior to and every day thereafter with FK506) or c) transiently immunosuppressed (treated with 10 mg/kg of FK506 orally and 1 mg/kg dexamethasone subcutaneously one day prior to, one hour prior to and one day after intraarterial delivery of pDNA) (Table 2). In untreated rats, luciferase expression was lost after 7 days from the CMV promoter or after 21 days from the MCK promoter. In either pCI-Luc+ or pMI-Luc+ injected rats, anti-luciferase antibodies were detected using ELISA by day 21 and were present at higher levels at day 56 and 70 after intravascular pDNA delivery (data not shown).
    TABLE 2
    Time course of luciferase expression (ng/g muscle) in hind limbs
    following intraarterial injections with 500 μg of pCI-Luc+ (A)
    or pMI-Luc+ (B) into rats treated with various immunosuppression
    regimens.
    Time CONDITION
    After Injection No Transient Continuous
    (Days) Treatment Immunosuppression Immunosuppression
    A. pCI-Luc+
    2 990.9
    7 492.6
    21 22.1
    30 10.3 672.0 1212.0
    56 0.3
    70 0.1 17.3 464.0
    B. pMI-Luc+
    2 37.3
    7 499.9
    21 286.9
    30 1260.0
    56 3.3
    70 0.3 571.0 1140.0
  • Example 6
  • Repetitive Injections. Sprague-Dawley rats (150 g) were injected intra-arterially in the right leg using 500 μg of pCI-Luc+ under increased pressure conditions on day 0. On days 7 and 14 the rats were injected slowly with 300 μg pCI-Luc+ in 1 ml into the tail vein. On day 24, the left leg was injected intraarterially with 500 μg of pCI-Luc+. On day 26, the animals were sacrificed and the left leg revealed luciferase expression (mean=4,500 ng of total luciferase/leg muscles, n=2) similar to the levels achieved in animals not pre-injected with pDNA (mean=6,940 ng/leg muscles, n=26).
  • In order to explore the ability to access different populations of myofibers, the same leg in rats were injected with the 500 μg of the β-galactosidase vector (pCI-LacZ) and two days hence with 500 μg of the nuclear GFP vector (pEBFP-N1). At two days after the last injection, the muscles were analyzed for expression of the two reporter genes. Expression of GFP and β-galactosidase was most often located in different myofibers (FIGS. 2A and C), but in some cells expression was coincident (FIG. 2B).
  • Example 7
  • Labeled pDNA Distribution in Muscle. Rhodamine-labeled pDNA (Rh-pDNA) was injected into the femoral artery of rats under various conditions in order to explore the uptake mechanism in muscle as was done for liver. When the injections were performed without impeding blood outflow (low intravascular pressure), almost no DNA was detected within the muscle tissues or vessels. FIG. 3A presents a rare field when some DNA can be seen between muscle cells. When the injections were performed with outflow occlusion (increased intravascular pressure), Rh-pDNA was detected throughout all the muscle (FIGS. 3B and 3C). At 5 min after injection, examination of tissue sections indicated that the majority of the Rh-pDNA was surrounding the muscle cells and there was no intracellular staining (FIG. 3B, arrow). At one hour after injection, substantial amounts of DNA can be seen inside the cells (FIG. 3C, arrowhead). Examination of serial confocal sections indicates that the intracellular staining pattern is punctate, unlikely consistent with a T tubular distribution.
  • Example 8
  • Expression of a therapeutic gene in skeletal muscle tissue. A plasmid DNA (pCI-hF9) expressing the human factor IX gene (cDNA) under transcriptional control of the human cytomegalovirus promoter was delivered to rat hind limb skeletal muscle. A midline abdominal incision was made and skin flaps were folded away with clamps to expose target area. Intestines were moved to visualize the iliac veins and arteries. Microvessel clips were placed on the external iliac, caudal epigastric, internal iliac, deferent duct, and gluteal arteries and veins to block both outflow and inflow of the blood to the leg. An efflux enhancer solution (0.5 mg papaverine in 3 ml saline) was injected into the external iliac artery though a 25 g needle, followed by the pDNA containing solution (500 μg in 10 ml Ringer's) 5 minutes later. The pDNA solution was injected in approximately 10 seconds. The microvessel clips were removed 2 minutes after the injection, and the peritoneum and skin were closed using sutures. The rats were immunosuppressed by treatment with 10 mg/kg of FK506 orally and 1 mg/kg dexamethasone subcutaneously one day prior to, one hour prior to, and one day after plasmid DNA delivery The rats were sacrificed after 3 weeks, at which time the hind limb skeletal muscles were removed and homogenized in a total volume of 60 ml. Human factor IX levels in the rat sera were determined using an ELISA and compared to normal human serum. Expression levels in 3 rats were 1400, 1000, and 1150 ng/ml extract, respectively. Therefore, the total amount of human factor IX present in the rat muscle tissue three weeks after pDNA delivery was approximately 70 μg.
  • Example 9
  • Expression of secreted alkaline phosphate from rat skeletal muscle cells. A plasmid DNA expression vector (pMIR54) was constructed in which the secreted alkaline phosphatase (SEAP) gene (obtained from plasmid pSEAP-2 basic, Clontech) is under transcriptional control of the human cytomegalovirus promoter. A solution of 500 μg pMIR54 in 10 ml Ringer's was injected into the iliac artery of Sprague Dawley rats as described in Examples above. The rats were immunosuppressed by treatment with 2.5 mg/kg of FK506 orally and 1 mg/kg dexamethasone subcutaneously one day prior to, one hour prior to plasmid DNA delivery. Following the pDNA delivery, the rats were treated with 2.5 mg/kg FK506 daily. Blood samples were obtained from these rats at several time points following plasmid DNA delivery. SEAP expression was determined using a chemiluminescent assay (Tropix) and compared to a standard curve.
    TABLE 3
    SEAP expression in rat muscle cells following DNA
    delivery.
    SEAP expression
    (ng SEAP per
    ml serum)
    Day 7 Day 14
    Rat 2889 2,301 1,407
    Rat 2992 3,735 2,942
  • Example 10
  • Expression in Multiple Muscle Groups. 500 μg of pCI-Luc in 10 ml of normal saline solution was injected into the femoral artery of adult rats in which a tourniquet was applied to the outside of the leg proximal (tourniquet was applied to the upper portion of the quadriceps group of muscles) to the injection site. Five days after injection, the different muscle groups from the leg were removed and cut into equal sections. Each section was placed into lysis buffer, the muscles were homogenized and 10 μl of the resulting lysates were assayed for luciferase activity.
  • High levels of luciferase expression were expressed in all muscle groups that were located distal to the tourniquet. These included the biceps femoris, posterior muscles of the upper leg, gastrocnemius, muscles of the lower leg, and muscles of the plantar surface.
    TABLE 4
    Luciferase expression in the various muscles of the rat leg after the
    injection of 500 μg of pCILuc into the femoral artery with a
    tourniquet applied around the outside of the upper leg muscles.
    Muscle Group Total Luciferase (ng/muscle group)
    Intravascular Delivery to Rat Leg (with external tourniquet)
    Upper leg anterior 0.181*
    (quadriceps) (* majority of this muscle group was
    above the tourniquet)
    Upper leg middle 28.3
    (biceps femoris)
    Upper leg posterior 146
    (hamstrings)
    Lower leg posterior 253.6
    (gastrocnemius)
    Lower leg anterior 115.2
    (lower shin muscles)
    Muscles of the plantar 0.433
    surface
    Intravascular Delivery to Rat Leg (without tourniquet)
    Upper leg anterior 0.010
    (quadriceps)
    Upper leg middle 0.011
    (biceps femoris)
    Upper leg posterior 2.16
    (hamstrings)
    Lower leg posterior 1.57
    (gastrocnemius)
    Lower leg anterior 0.72
    (lower shin muscles)
    Muscles of the plantar 0.202
    surface
  • Intravascularly-administered plasmid DNA is expressed efficiently in multiple muscle groups when blood flow is impeded using an external tourniquet.
  • Example 11
  • Delivery of Polynucleotides to Limb Skeletal Muscle in Mdx Mice. ICR or mdx mice, ˜30 gram, were anesthetized by intramuscular injection of ketamine (80-100 mg/kg) and xylazine (2 mg/kg). Metofane was added through inhalation if necessary during the procedure. A median incision was made from the upper third of abdomen to the caudal edge of the abdominal and the right caudal part of abdominal cavity was exposed using retractors. The tissue in front of the right external iliac artery was cleaned by forceps and a cotton tipped applicator. The arteries and veins to be clamped were separated from surrounding tissue and the caudal epigastric artery and vein, internal iliac arteries and vein, gluteal artery and vein, the vessels of deferent duct and external iliac artery and vein were clamped. A 0.6 ml of papaverine solution (containing 0.1 mg of papaverine) was injected into external iliac artery distal to the clamp. 2.5-3 ml of DNA solution containing 100 μg plasmid DNA was injected into the external iliac artery distal to the clamp with pressure 5 minutes post papaverine injection. A piece of gelfoam was put on the injection site before withdrawal of the needle and pressure was kept on the gelfoam to prevent bleeding. The clamps are taken off 2 minutes after injection and the abdominal cavity was closed by suturing.
  • Muscle samples were taken 7-10 days after injection and 6 μm thickness cryostat sections were made. Endogenous peroxidase activities were blocked by incubating the sections in 0.3% hydrogen peroxide in PBS for 5-10 min after the sections were mounted on slides and dried. The sections were rinsed twice with PBS (2 min×2) followed by Avidin/Biotin blocking by using Vector Avidin/Biotin Blocking Kit (Cat. No. SP-2001). The following steps were done according to the procedure of Vector M.O.M Immunodetection Kit (Cat. No. PK-2200). The immunofluorescent staining for human dystrophin in mouse muscle was done following the procedure of Vector M.O.M Immunodetection Kit (Cat. No. FMK-2201). For luciferase assays, 5 groups of muscle were taken from the whole injected leg according to their distribution, the anterior, posterior, medial, anterior of low leg and posterior of low leg. 2 ml of cold lysis buffer was added to each group of muscle followed by homogenization. Luciferase activity was measured by luminometer and the light units were converted to luciferase protein by using the converting rate of pg=light units×solution volume/20×2.05/100,000
  • Results are shown in Table 5 and FIG. 4. FIG. 4A shows β-galactosidase expression in mdx dystrophic mouse. FIGS. 4 B and C show human dystrophin expression in leg skeletal muscle in mdx and normal mouse, respectively.
    TABLE 5
    Luciferase expression in Mdx mouse.
    total expression of whole leg expression of per gram muscle
    animal # (ng luciferase) (ng luciferase/gram muscle)
    134 1691 1271
    135 1738 1307
    137 1248 1177
    138 869 643
    140 1641 1357
    141 881 663
    average 1345 1070
  • These data demonstrate that the nucleic acid delivery process results in expression of luciferase, β-galactosidase and the therapeutic protein dystrophin in both normal and dystrophic mice. Furthermore, expression of luciferase and β-galactosidase correlate with expression of dystrophin.
  • Example 12
  • Intravenous delivery of the therapeutically relevant dystrophin gene to limb muscle cells in mouse. A clinically-relevant example is provided by the intravenous delivery of the human dystrophin gene into the mdx4cv mouse model for Duchenne muscular dystrophy, the dystrophin-negative strain B6Ros.Cg-DMDmdx-4Cv (Jackson laboratory). For each injection, 300 μg of a pDNA human dystrophin expression vector (Acsadi et al. 1991) in 0.6 ml of NSS (7.5 s injection) was injected into a distal site in the great saphenous vein of the mouse hind limb. Fluid flow into and out of the leg was occluded by means of a tourniquet. Blood flow was occluded prior to injection and for two minutes following the injection. Immunohistochemical staining for human dystrophin expression in mdx4cv mouse muscle (from gastrocnemius) was performed one week post-injection using a mouse, anti-dystrophin polyclonal primary antibody and a FITC-conjugated goat, anti-mouse IgG (FAB specific; Sigma) secondary antibody. Similar percentages of dystrophin-positive myofibers were detected using a monoclonal antibody specific for human dystrophin (NCL-DYS3, Novocastra Laboratories). Images were captured using a 10× objective (Zeiss Axioplan 2 fluorescent microscope).
  • In four mdx4cv mice injected once intravenously with a plasmid expression vector encoding full-length, human dystrophin, 3-15% of myofibers of various hind limb muscles exhibited sarcolemmal dystrophin expression (FIG. 5). Dystrophin-positive revertants in this particular mdx strain are below 0.5% (FIG. 5). The ability to perform the intravenous procedure in mouse models enhances its utility as a research tool.
  • Example 13
  • Delivery of Polynucleotides to Limb Skeletal Muscles in Dystrophic Dog Model. Juvenile male Golden Retriever dogs of 3 to 12 kg body weight underwent intra-arterial injections in their limbs following anesthesia. Anesthesia was with intravascular injection of propofol followed by isoflurane inhalant. For forearm injections, the arm was put at the extension and external rotation position and a 3 cm incision was made at the conjunction of armpit and upper arm and near the inside edge of the brachial biceps. After separating the brachial artery from the brachial vein and median nerve, a catheter (3-4F) was inserted anterograde into the brachial artery until the tip of the catheter reached to the elbow and was fixed by ligation. In some cases the brachial vein was clamped. Blood circulation of the forelimb was further inhibited by using a tourniquet placed around the upper limb up to the elbow (10 minutes maximum). For whole hind leg injections, an incision was made through the midline of the abdomen one inch below the umbilicus to the pubis. Connective tissue was separated to expose the common iliac artery and vein, external iliac artery and vein, internal iliac artery and vein, inferior epigastric artery and vein, superficial epigastric artery and vein, and the superficial iliac circumflex artery and vein. Clamps were placed on the inferior epigastric artery and vein, superficial epigastric artery and vein, and the superficial iliac circumflex artery and vein. A catheter (F5) was placed into the distal part of the iliac artery to the femoral artery and secured by ligation at the beginning of the femoral artery. Clamps are then placed on the external iliac vein, internal iliac artery and vein, and the common iliac artery and vein.
  • A 17% papaverine/saline solution was injected to increase vessel dilation (10-50 ml depending on animal size). After 5 minutes a plasmid DNA/saline solution was injected at moderately increased pressure using a nitrogen-pressurized cylinder set at 65 psi. For the forelimbs, the injection volume was 50-200 ml. For whole leg injections, the injection volume was 60-500 ml. Injection rates varied from 20 s to 120 s. Two min after injection, the clamps and tourniquet were released and the catheters were removed.
  • One forelimb and the opposite hind limb or all four limbs were injected on day one with pMI-Luc+ (20-50 mg) or the dystrophin plasmid (50-330 mg). In these vectors, the reporter genes are under transcriptional control of the muscle creatine kinase promoter, which has been shown to direct sustained, high level expression in muscle. The animals were sacrificed at 7 days and all muscles were analyzed for gene expression. Uninjected limbs or limbs injected with saline were used to test for revertants. Results are shown in Table 6 and graphically summarized in FIG. 6. FIG. 6A illustrates the distribution of luciferase expression in normal dog. FIG. 6B illustrates the distribution of luciferase expression in the dystrophic dog model.
    TABLE 6
    Luciferase expression after of delivery pCI-Luc polynucleotide in dog
    skeletal muscle cells. Numbers given in pg Luciferase per mg total protein.
    GRMD dog healthy dog
    left right left right
    antebrachial muscles
    dorsolateral extensor carpi radialis 0.8 633
    extensor digitorium communis 5 1570 299
    extensor digitorium lateralis 7915 438.5
    extensor carpi ulnaris 671 21.5
    extensor pollicis longus et indicis proprius 6763 2456.7
    abductor pollicis longus 16724 292.4
    supinator 9 14395 3.3 1920.8
    caudal flexor carpi radialis 3 828 1.5 116.2
    flexor carpi ulnaris 270 6.1
    flexor digitorum superficialis 2017 43.5
    flexor digitorum profundus 49 11.3
    pronator teres 9231 5.2 270.6
    forepaw forepaw 10 958 2 1048.7
    other brachi radialis 545.1
    muscles of the crus
    craniolateral tibialis cranialis 980 1.4 1.7
    extensor digitorum longus 992 0.3
    peroneus longus 4116 0.3 127.8
    peroneus brevis 6.2
    extensor digitorum lateralis 0.2
    caudal gastrocnemius 4365 0.1 3 0.1
    flexor digitorum profundus 1912 1.9 3
    tibialis caudalis 0.4
    popliteus 9821 0.3
    other Testes 0.1
    Liver #1 0.3
    muscles of the pelvic limb
    thigh gluteus superficialis 1.4 4.9
    gluteus medius 4 0.2 0.1
    sartorius 661.2
    tensor fasciae latae 0.5 369.7
    biceps femoris 10312 1.1 0.1
    0.6
    semimembranosus 5988 1.7 49.8
    semitendinosus 432 1.1 0.1
    abductor magnus brevis 4103 2 3644.8
    sartorius cranial part 4664 0.9
    rectus femoris 396 0.1 179.9
    vastus medialis 2588 0.5 7.4
    vastus intermedius 4469 3.2 12448.7
    vastus lateralis 2102 1 2927.8
    pectineus 737 0.1 11.9
    gracilis 1826 0.5 146
    gluteal region piriformis 14 1.2
    and gemellus 3
    hip joint quadratus femoris 911 0.1 1
    gluteus profundus 0 1.8
    PR21?obturator externus 1.8
    biceps brachialis 0.1
  • Example 14
  • Delivery of PEI/DNA, Histone H1/DNA, and MC66/DNA Particles to Multiple Skeletal Muscles in Rat Via a Single Injection into a Blood Vessel. PEI/DNA and histone H1/DNA particles were injected into rat leg muscle by a single intra-arterial injection into the external iliac (Budker et al. 1998). For MC66/DNA particles, 500 μg pDNA (500 μl) was mixed with M66 copolymer at a 1:3 wt:wt ratio in 500 μl saline. Complexes were then diluted in Ringers solution to total volume of 10 mls. Female Harlan Sprague Dawley (HSD SD) rats, approximately 150 g, were used for the muscle injections. Each received complexes containing 100 μg plasmid DNA encoding the luciferase gene under control of the CMV enhancer/promoter (pCI-Luc) (Zhang et al. 1997). A total volume of 10 mls was injected into the iliac artery of Sprague-Dawley rats in approximately 10 seconds.
  • Animals were sacrificed after 1 week and individual muscle groups were removed and assayed for luciferase expression. Results of the rat injections are provided in relative light units (RLUs) and micrograms (μg) of luciferase produced. To determine RLUs, 10 μl of cell lysate were assayed luminometer and total muscle RLUs were determined by multiplying by the appropriate dilution factor. To determine the total amount of luciferase expressed per muscle we used a conversion equation that was determined in an earlier study (Zhang et al. 1997) (pg luciferase=RLUs×5.1×10−5)
    A. DNA/PEI particles (1:0.5 charge ratio)
    Total Total
    Muscle Group RLUs Luciferase
    muscle group 1 (upper leg anterior) 3.50 × 109 0.180 μg
    muscle group 2 (upper leg posterior) 3.96 × 109 0.202 μg
    muscle group 3 (upper leg medial) 7.20 × 109 0.368 μg
    muscle group 4 (lower leg posterior) 9.90 × 109 0.505 μg
    muscle group 5 (lower leg anterior) 9.47 × 108 0.048 μg
    muscle group 6 (foot) 6.72 × 106 0.0003 μg 
  • B. DNA/histone H1 particles (1:0.5 charge ratio)
    Total Total
    Muscle Group RLUs Luciferase
    muscle group 1 (upper leg anterior) 3.12 × 109 0.180 μg
    muscle group 2 (upper leg posterior) 9.13 × 109 0.202 μg
    muscle group 3 (upper leg medial) 1.23 × 1010 0.368 μg
    muscle group 4 (lower leg posterior) 5.73 × 109 0.505 μg
    muscle group 5 (lower leg anterior) 4.81 × 108 0.048 μg
    muscle group 6 (foot) 6.49 × 106 0.0003 μg 
  • C. DNA/M66 polymer particles
    Total Total
    Muscle Group RLUs Luciferase
    muscle group 1 (upper leg anterior) 3.58 × 109 0.183 μg
    muscle group 2 (upper leg posterior) 6.46 × 108 0.032 μg
    muscle group 3 (upper leg medial) 2.63 × 109 0.134 μg
    muscle group 4 (lower leg posterior) 1.97 × 109 0.101 μg
    muscle group 5 (lower leg anterior) 3.19 × 109 0.163 μg
  • Results indicated high level luciferase expression throughout the leg with a single injection of PEI/DNA, histone H1/DNA, or MC66/DNA particles.
  • Example 15
  • Increased vascularization following delivery of a therapeutic polynucleotide to primate limb. DNA delivery was performed via brachial artery with blood flow blocked by a sphygmomanometer cuff proximately to the injection site. Left arm was transfected with VEGF, while right arm was transfected with EPO. The Sartorious muscle from left leg was used as non-injected control. A male Rhesus monkey weighing 14 kg was used for these injections. The animal was anesthetized with Ketamine (10-15 mg/kg). A modified pediatric blood pressure cuff was positioned on the upper arm. The brachial artery was cannulated with a 4 F angiography catheter. The catheter was advanced so that the tip was positioned just below the blood pressure cuff. Prior to the injection, the blood pressure cuff was inflated so that the cuff pressure was at least 20 mmHg higher than the systolic blood pressure. After cuff inflation, papaverine (5 mg in 30 ml of saline) was injected by hand (˜8 to 10 seconds). After 5 min, the pDNA solution was delivered rapidly with a high volume injection system. For the EPO injection, 10 mg of pDNA was added to 170 ml of saline and injected at a rate of 6.8 ml per second. For the VEGF injection, 10 mg of pDNA was added to 150 ml of saline, and injected at a rate of 5.4 ml per second.
  • After 65 days, the animal was euthanized by overdose I.V. injection of pentobarbital Ketamine (10 mg/kg). The entire Pronator quadratus and Pronator teres muscles from both sides were immediately harvested and fixed for 3 day in 10% neutral buffered formalin (VWR, Cleveland, Ohio). After fixation, an identical grossing was performed for left and right muscles and slices across the longitudinal muscles were taken. Specimens were routinely processed and embedded into paraffin (Sherwood Medical, St. Louis, Mo.). Four microns sections were mounted onto precleaned slides, and stained with hematoxylin and eosin (Surgipath, Richmond, Ill.) for pathological evaluation. Sections were examined under Axioplan-2 microscope and pictures were taken with the aid of AxioCam digital camera (both from Carl Zeiss, Goettingen, Germany).
  • To evaluate the effect of VEGF plasmid delivery on cell composition in muscle tissue and neo-angiogenesis, we used monoclonal mouse anti-human CD31 antibody (DAKO Corporation, Carpinteria Calif.). The immunostaining was performed using a standard protocol for paraffin sections. Briefly: four microns paraffin sections were deparaffinized and re-hydrated. Antigen retrieval was performed with DAKO Target Retrieval Solution (DAKO Corporation, Carpinteria Calif.) for 20 min at 97° C. To reduce non-specific binding the section were incubated in PBS containing 1% (wt/vol) BSA for 20 min at RT. Primary antibody 1:30 in PBS/BSA were applied for 30 min at RT. CD31 antibody were visualized with donkey anti-mouse Cy3-conjugated IgG, 1:400 (Jackson Immunoresearch Lab, West Grove Pa.) for 1 h at RT. ToPro-3 (Molecular Probes Inc.) was used for nuclei staining; 1:70,000 dilution incubated for 15 min at RT. Sections were mounted with Vectashield non-fluorescent mounting medium and examined under confocal Zeiss LSM 510 microscope (Carl Zeiss, Goettingen, Germany). Images were collected randomly under 400× magnification, each image representing 0.106 sq mm. Because muscle fibers and red blood cells have an autofluorescence in FITC channel we use a 488 nm laser to visualize these structures. Morphometry analysis. Coded mages were opened in Adobe Photoshop 5.5 having image size 7×7 inches in 1×7 inches window, and a grid with rulers was overlaid. The number of muscle fibers, CD31 positive cells and total nuclei was counted in all 7 image's strips consecutively, without any knowledge of experimental design. T-Test for Two-Sample Unequal Variances was used for statistical analysis.
  • Results: Microscopic evaluation did not reveal any notable pathology in either muscle regardless of the gene delivered. Also, neither muscle showed any notable presence of inflammatory cells, except of few macrophages. Necrosis of single muscle fibers was extremely rare in both, occupying negligible volume and was not associated with infiltration/vascularization. However, in muscles transfected with VEGF-165 plasmid, the interstitial cell and vascular density (observed in H&E-stained slides) was obviously increased (FIG. 7), as compare to EPO plasmid administered muscle (FIG. 7). Based on morphologic evaluation, these newly arrived interstitial cells we suggested to be endothelial and adventitial cells, smooth muscle cells, and fibroblasts. To evaluate participation of endothelial cells in this neo-morphogenesis, we have counted the number of CD31 positive cells in EPO and VEGF delivered Pronator quadratus muscles (FIG. 8). To assure that comparable specimens were analyzed in right and left muscles, the number of muscle fibers was counted per area unit (0.106 sq mm). The VEGF and EPO administered muscles were not different in muscle fiber number (means 30.5 and 31.6). The number of CD31 positive cells however was significantly increased by 61.7% p<0.001 (means 53.2 vs. 32.9).
  • Example 16
  • Intravascular Delivery of Non-viral Complexes to Skeletal Muscle Cells. All particles were injected into rat leg muscle via a single intra-arterial injection into the external iliac (Budker et al. 1998) artery of Harlan Sprague Dawley (HSD SD) rats. All rats used were female and approximately 150 g and each received complexes containing 100 μg of plasmid DNA encoding the luciferase gene under control of the CMV enhancer/promoter (pCI-Luc).
  • Results of the rat injections are in relative light units (RLUs) and μg of luciferase produced. To determine RLUs, 10 μl of cell lysate were assayed using an EG&G Berthold LB9507 luminometer and total muscle RLUs were determined by multiplying by the appropriate dilution factor. To determine the total amount of luciferase expressed per muscle we used a conversion equation that was determined in an earlier study (Zhang et al. 1997) (ng luciferase=RLUs×5.1×10−8).
  • Results: For this study a variety of DNA/polycation complexes were formulated using three different charge ratios such that the net charge of complexes was either negative (two formulations) or positive (one formulation). Polycations used in this study included; proteins, polymers, lipids, polyamines, and combinations of each. In all cases the negatively charged complexes resulted in higher levels of gene expression in rat muscle following delivery than the positively charged complexes (see table 7).
    TABLE 7
    Luciferase expression in rat leg muscles following injection
    into the iliac artery of negative vs. positive charge DNA complexes.
    Total nanograms of
    Charge Ratio luciferase expression
    (negative to per limb (all muscle
    DNA/Polycation Complex positive) groups combined)
    Polynucleotide alone 1:0 1369.9
    DNA/cationic protein 1:0.25 1908.8
    (histone H1) 1:0.5 135.2
    1:2 69.1
    DNA/cationic polymer 1:0.25 2355.3
    (linear PEI) 1:0.5 1677.9
    1:2 7.2
    DNA/cationic protein + polyamine 1:0.25 1551.1
    (histone H1 + polyamine 58) 1:0.5 1181.9
    1:2 16.6
    DNA/cationic lipid 1:0.25 537.3
    (DOTAP) 1:0.5 171.6
    1:2 1.8
    DNA/cationic protein + cationic 1:0.25 863.4
    lipid 1:0.5 286.0
    (histone H1 + DOTAP) 1:2 7.5
  • Conclusions: When using the bloodstream for gene delivery to skeletal muscle cells, the net charge of the complex is very important. Regardless of the type of polycation used for complexation with the DNA, net negatively charged complexes are much more efficient for gene delivery and expression than positively charged complexes.
  • Example 17
  • Delivery of polynucleotide to the diaphragm. The monkey was anesthetized with ketamine followed by halothane inhalation. A 2 cm long incision was made in the upper thigh close to the inguinal ligament just in front of the femoral artery. Two clamps were placed around the femoral vein after separating the femoral vein from surrounding tissue. At an upstream location, the femoral vein was ligated by the clamp and a guide tube was inserted into the femoral vein anterogradely. A French 5 balloon catheter (D 1.66 mm) with guide wire was inserted into the inferior vena cava through the guide tube and an X-ray monitor was used for instructing the direction of guide wire. The guide wire was directed into the inferior phrenic vein. The catheter position in the inferior phrenic vein was checked by injecting iodine. The balloon was inflated to block blood flow through the inferior phrenic vein. 20 ml 0.017% papaverine in normal saline was injected. 5 minutes after papaverine injection, 40 ml of DNA solution (3 mg) was injected under elevated pressure (65 sec injection time). 2 min after DNA injection, the balloon was released and the catheter was removed. The animal was sacrificed and the diaphragm was taken for luciferase assay 7 days after the procedure.
    TABLE 8
    Luciferase expression in diaphragm from monkey
    sacrificed 7 days after injection of pCI-Luc+.
    total luciferase ng luciferase/
    diaphragm section (ng) gram if tissue
    anterior part of left side 0 0
    posterior part of left side 0 0
    left conjunction area 0 0
    anterior part of right side 221.94 27.88
    posterior part of right side 15.98 2.12
    right conjunction area 34.21 17.82
  • Example 18
  • Co-administration of a vasoconstrictor. M-methyl-L-arginine (L-NMMA) is a competitive inhibitor of nitric oxide synthetase. pCI-Luc was injected into the iliac artery of rat following a short pre-treatment with L-NMMA. A 4 cm long abdominal midline excision was performed in 150-200 g, adult Sprague-Dawley rats anesthetized with 80 mg/mg ketamine and 40 mg/kg xylazine. Microvessel clips were placed on external iliac, caudal epigastric, internal iliac and deferent duct arteries and veins to block both outflow and inflow of the blood to the leg. 3 ml of normal saline with 0.66 mM L-NMMA were injected into the external iliac artery. After 2 min, a 27 g butterfly needle was inserted into the external iliac artery and 10 ml of DNA solution (50 μg/ml pCI-Luc) in normal saline was injected within 8-9 sec. Luciferase assays were performed on limb muscle samples (quadriceps femoris) 2 days after injection. Luciferase expression was determined as previously reported (Wolff et al. 1990) A LUMAT™ LB 9507 (EG&G Berthold, Bad-Wildbad, Germany) luminometer was used.
    TABLE 9
    Luciferase expression in rat leg muscle following injection of DNA
    into the iliac artery. Enhancement of delivery with
    M-methyl-L-arginine.
    Total Luciferase
    Organ Treatment (nanograms)
    Muscle (quadriceps) +papaverine  9,999
    Muscle (quadriceps) +0.66 mM L-NMMA 15,398
    Muscle (quadriceps) +papaverine, 24,829
    +0.66 mM L-NMMA
  • The foregoing is considered as illustrative only of the principles of the invention. Furthermore, since numerous modifications and changes will readily occur to those skilled in the art, it is not desired to limit the invention to the exact construction and operation shown and described. Therefore, all suitable modifications and equivalents fall within the scope of the invention.

Claims (17)

1. An in vivo process for delivering a polynucleotide to limb muscle cell in a mammal affected by Muscular Dystrophy comprising:
a) inserting an injector into a blood vessel in the limb of the mammal;
b) applying a device external to mammalian skin for compressing vessels in an area underneath the cuff thereby occluding fluid flow into and out of the limb; and,
c) injecting a solution containing the polynucleotide into the lumen of said blood vessel distal to the occlusion wherein the solution is retained within the limb, thereby increasing the volume of fluid in the limb and delivering the polynucleotides to skeletal muscle cells in the limb.
2. The process of claim 1 wherein the polynucleotide is selected from the group consisting of viral vector and non-viral vector.
3. The process of claim 1 wherein the externally applied device consists of a tourniquet placed over the skin.
4. The process of claim 1 wherein the externally applied device consists of a cuff placed over the skin.
5. The process of claim 4 wherein the externally applied device consists of a sphygmomanometer cuff placed over the skin.
6. The process of claim 1 wherein the polynucleotide is expressed.
7. The process of claim 6 wherein the polynucleotide encodes a peptide or protein.
8. The process of claim 7 wherein the peptide or protein is secreted from the muscle cell.
9. The process of claim 7 wherein the peptide or protein provides a therapeutic benefit to the mammal.
10. The process of claim 7 wherein the polynucleotide encodes a gene selected from the group consisting of: dystrophin, dystrophin associated glycoprotein, sarcoglycan complex protein, β-sarcoglycan, δ-sarcoglycan, α-sarcoglycan, γ-sarcoglycan, utrophin, calpain, caveolin-3, laminin-α2, fukutin, emerin, myotilin, lamin A/C, calpain-3, dysferlin, and telethonin.
11. The process of claim 7 wherein the peptide or protein alleviates secondary defects of Muscular Dystrophy.
12. The process of claim 11 wherein the peptide or protein is selected from the group consisting of: mini-agrin, laminin α2, α7 integrin, GalNac transferase, ADAM12 protein, calpastatin, nitric oxide synthase, IGF-I, dominant negative myostatin, myostatin inhibitors, TGF-β, actin, titin, muscle creatine kinase, troponin, human growth factor, human growth hormone releasing hormone, vascular endothelial growth factor, insulin, and anti-inflammatory genes.
13. The process of claim 1 wherein the mammal has Duchenne Muscular Dystrophy, Becker Muscular Dystrophy, autosomal recessive limb-girdle Muscular Dystrophy, autosomal-dominant limb-girdle Muscular Dystrophy, merosin-deficient congenital Muscular Dystrophy, Fukuyama type congenital Muscular Dystrophy, or Emery-Dreifuss Muscular Dystrophy.
14. The process of claim 1 wherein the polynucleotide inhibits expression of an endogenous gene.
15. The process of claim 14 wherein the polynucleotide is selected from the group consisting of antisense oligonucleotide, siRNA, miRNA, hairpin RNA, or RNAi inducing oligonucleotide.
16. The process of claim 14 wherein the polynucleotide expresses an antisense oligonucleotide, double strand oligonucleotide, siRNA, miRNA, and RNAi inducing oligonucleotide
17. The process of claim 14 wherein the mammal is affected by an autosomal dominant Muscular Dystrophy.
US11/694,140 1999-11-05 2007-03-30 Devices and processes for distribution of genetic material to mammalian limb Expired - Fee Related US7642248B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/694,140 US7642248B2 (en) 1999-11-05 2007-03-30 Devices and processes for distribution of genetic material to mammalian limb

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US16371999P 1999-11-05 1999-11-05
US09/707,117 US7396821B1 (en) 1999-11-05 2000-11-06 Intravascular delivery of nucleic acid
US09/707,000 US7507722B1 (en) 1999-11-05 2000-11-06 Intravascular delivery of nucleic acid
US10/339,934 US20070173465A9 (en) 1995-10-11 2003-01-10 Expression of zeta negative and zeta positive nucleic acids using a dystrophin gene
US10/429,478 US7214369B2 (en) 2003-05-05 2003-05-05 Devices and processes for distribution of genetic material to mammalian limb
US11/694,140 US7642248B2 (en) 1999-11-05 2007-03-30 Devices and processes for distribution of genetic material to mammalian limb

Related Parent Applications (4)

Application Number Title Priority Date Filing Date
US09/707,117 Continuation-In-Part US7396821B1 (en) 1995-12-13 2000-11-06 Intravascular delivery of nucleic acid
US09/707,000 Continuation-In-Part US7507722B1 (en) 1995-12-13 2000-11-06 Intravascular delivery of nucleic acid
US10/339,934 Continuation-In-Part US20070173465A9 (en) 1995-10-11 2003-01-10 Expression of zeta negative and zeta positive nucleic acids using a dystrophin gene
US10/429,478 Continuation-In-Part US7214369B2 (en) 1999-11-05 2003-05-05 Devices and processes for distribution of genetic material to mammalian limb

Publications (2)

Publication Number Publication Date
US20070244067A1 true US20070244067A1 (en) 2007-10-18
US7642248B2 US7642248B2 (en) 2010-01-05

Family

ID=38605539

Family Applications (1)

Application Number Title Priority Date Filing Date
US11/694,140 Expired - Fee Related US7642248B2 (en) 1999-11-05 2007-03-30 Devices and processes for distribution of genetic material to mammalian limb

Country Status (1)

Country Link
US (1) US7642248B2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010125358A1 (en) * 2009-04-30 2010-11-04 The University Of Birmingham Method for diagnosing and treating muscular dystrophy
US20100323001A1 (en) * 2006-11-08 2010-12-23 Veritas Llc In Vivo Delivery Of Double Stranded RNA To a Target Cell
CN102807993A (en) * 2012-08-31 2012-12-05 西北农林科技大学 Skeletal muscle specific miRNA expression vector and reconstitution cell of target Myostatin gene
WO2012145597A3 (en) * 2011-04-21 2012-12-27 Nationwide Children's Hospital, Inc. Recombinant virus products and methods for inhibition of expression of myotilin
WO2017048732A1 (en) * 2015-09-14 2017-03-23 Research Institute At Nationwide Children's Hospital Recombinant virus products and methods for inhibition of expression of myotilin
US11091760B2 (en) 2011-04-21 2021-08-17 Nationwide Children's Hospital, Inc. Recombinant virus products and methods for inhibition of expression of myotilin

Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5521291A (en) * 1991-09-30 1996-05-28 Boehringer Ingelheim International, Gmbh Conjugates for introducing nucleic acid into higher eucaryotic cells
US5580859A (en) * 1989-03-21 1996-12-03 Vical Incorporated Delivery of exogenous DNA sequences in a mammal
US5583020A (en) * 1992-11-24 1996-12-10 Ribozyme Pharmaceuticals, Inc. Permeability enhancers for negatively charged polynucleotides
US5698531A (en) * 1989-03-31 1997-12-16 The Regents Of The University Of Michigan Treatment of diseases by site-specific instillation of cells or site-specific transformation of cells and kits therefor
US5744335A (en) * 1995-09-19 1998-04-28 Mirus Corporation Process of transfecting a cell with a polynucleotide mixed with an amphipathic compound and a DNA-binding protein
US5922687A (en) * 1995-05-04 1999-07-13 Board Of Trustees Of The Leland Stanford Junior University Intracellular delivery of nucleic acids using pressure
US6177403B1 (en) * 1996-10-21 2001-01-23 The Trustees Of The University Of Pennsylvania Compositions, methods, and apparatus for delivery of a macromolecular assembly to an extravascular tissue of an animal
US6495131B1 (en) * 1997-07-11 2002-12-17 Crucell Holland B.V. Interleukin-3 gene therapy for cancer
US20030087867A1 (en) * 1999-07-09 2003-05-08 Vogels Ronald V. Gene therapy for enhancing and/or inducing angiogenesis
US6627616B2 (en) * 1995-12-13 2003-09-30 Mirus Corporation Intravascular delivery of non-viral nucleic acid
US6673039B1 (en) * 1997-12-19 2004-01-06 Trustees Of The University Of Pennsylvania Compositions, kits, methods, and apparatus for transvascular delivery of a composition to an extravascular tissue of a mammal
US20040023850A1 (en) * 2002-07-26 2004-02-05 Wolff Jon A. Delivery of molecules and complexes to mammalian cells in vivo
US20040072785A1 (en) * 1999-11-23 2004-04-15 Wolff Jon A. Intravascular delivery of non-viral nucleic acid
US20040136960A1 (en) * 2003-01-10 2004-07-15 Wolff Jon A. Devices and processes for distribution of genetic material to mammalian limb
US20040224879A1 (en) * 2003-05-05 2004-11-11 Wolff Jon A. Devices and processes for distribution of genetic material to mammalian limb
US20040242528A1 (en) * 2003-05-28 2004-12-02 Hagstrom James E. Intravenous delivery of polynucleotides to cells in mammalian limb
US20040259828A1 (en) * 1995-12-13 2004-12-23 Wolff Jon A. Intravascular delivery of non-viral nucleic acid
US20050119165A1 (en) * 1999-05-20 2005-06-02 Scios, Inc. Vascular endothelial growth factor dimers

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1246649B1 (en) 1999-11-05 2006-10-18 Mirus Bio Corporation Intravascular delivery of nucleic acid

Patent Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5580859A (en) * 1989-03-21 1996-12-03 Vical Incorporated Delivery of exogenous DNA sequences in a mammal
US5698531A (en) * 1989-03-31 1997-12-16 The Regents Of The University Of Michigan Treatment of diseases by site-specific instillation of cells or site-specific transformation of cells and kits therefor
US5521291A (en) * 1991-09-30 1996-05-28 Boehringer Ingelheim International, Gmbh Conjugates for introducing nucleic acid into higher eucaryotic cells
US5583020A (en) * 1992-11-24 1996-12-10 Ribozyme Pharmaceuticals, Inc. Permeability enhancers for negatively charged polynucleotides
US5922687A (en) * 1995-05-04 1999-07-13 Board Of Trustees Of The Leland Stanford Junior University Intracellular delivery of nucleic acids using pressure
US5744335A (en) * 1995-09-19 1998-04-28 Mirus Corporation Process of transfecting a cell with a polynucleotide mixed with an amphipathic compound and a DNA-binding protein
US20040259828A1 (en) * 1995-12-13 2004-12-23 Wolff Jon A. Intravascular delivery of non-viral nucleic acid
US6627616B2 (en) * 1995-12-13 2003-09-30 Mirus Corporation Intravascular delivery of non-viral nucleic acid
US6177403B1 (en) * 1996-10-21 2001-01-23 The Trustees Of The University Of Pennsylvania Compositions, methods, and apparatus for delivery of a macromolecular assembly to an extravascular tissue of an animal
US6495131B1 (en) * 1997-07-11 2002-12-17 Crucell Holland B.V. Interleukin-3 gene therapy for cancer
US6673039B1 (en) * 1997-12-19 2004-01-06 Trustees Of The University Of Pennsylvania Compositions, kits, methods, and apparatus for transvascular delivery of a composition to an extravascular tissue of a mammal
US20050119165A1 (en) * 1999-05-20 2005-06-02 Scios, Inc. Vascular endothelial growth factor dimers
US20030087867A1 (en) * 1999-07-09 2003-05-08 Vogels Ronald V. Gene therapy for enhancing and/or inducing angiogenesis
US20040072785A1 (en) * 1999-11-23 2004-04-15 Wolff Jon A. Intravascular delivery of non-viral nucleic acid
US20040023850A1 (en) * 2002-07-26 2004-02-05 Wolff Jon A. Delivery of molecules and complexes to mammalian cells in vivo
US20040136960A1 (en) * 2003-01-10 2004-07-15 Wolff Jon A. Devices and processes for distribution of genetic material to mammalian limb
US20040224879A1 (en) * 2003-05-05 2004-11-11 Wolff Jon A. Devices and processes for distribution of genetic material to mammalian limb
US20040242528A1 (en) * 2003-05-28 2004-12-02 Hagstrom James E. Intravenous delivery of polynucleotides to cells in mammalian limb

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100323001A1 (en) * 2006-11-08 2010-12-23 Veritas Llc In Vivo Delivery Of Double Stranded RNA To a Target Cell
EP2548438A1 (en) 2006-11-08 2013-01-23 Veritas Bio, LLC In vivo delivery of double stranded RNA to a target cell
US8524679B2 (en) 2006-11-08 2013-09-03 Veritas Bio, Llc In vivo delivery of double stranded RNA to a target cell
EP2799547A1 (en) 2006-11-08 2014-11-05 Veritas Bio, LLC In vivo delivery of RNA to a target cell
WO2010125358A1 (en) * 2009-04-30 2010-11-04 The University Of Birmingham Method for diagnosing and treating muscular dystrophy
WO2012145597A3 (en) * 2011-04-21 2012-12-27 Nationwide Children's Hospital, Inc. Recombinant virus products and methods for inhibition of expression of myotilin
US11091760B2 (en) 2011-04-21 2021-08-17 Nationwide Children's Hospital, Inc. Recombinant virus products and methods for inhibition of expression of myotilin
CN102807993A (en) * 2012-08-31 2012-12-05 西北农林科技大学 Skeletal muscle specific miRNA expression vector and reconstitution cell of target Myostatin gene
WO2017048732A1 (en) * 2015-09-14 2017-03-23 Research Institute At Nationwide Children's Hospital Recombinant virus products and methods for inhibition of expression of myotilin

Also Published As

Publication number Publication date
US7642248B2 (en) 2010-01-05

Similar Documents

Publication Publication Date Title
US7589059B2 (en) Delivery of molecules and complexes to mammalian cells in vivo
US6265387B1 (en) Process of delivering naked DNA into a hepatocyte via bile duct
US20090209630A1 (en) Gene delivery formulations and methods for treatment of ischemic conditions
US7473419B2 (en) Intravascular delivery of nucleic acid
US20030236214A1 (en) Charge reversal of polyion complexes and treatment of peripheral occlusive disease
US20040072785A1 (en) Intravascular delivery of non-viral nucleic acid
US7642248B2 (en) Devices and processes for distribution of genetic material to mammalian limb
US7214369B2 (en) Devices and processes for distribution of genetic material to mammalian limb
US7803782B2 (en) Intravenous delivery of polynucleotides to cells in mammalian limb
US20030130224A1 (en) Expression of zeta negative and zeta positive nucleic acids using a dystrophin gene
US20040136960A1 (en) Devices and processes for distribution of genetic material to mammalian limb
US7396821B1 (en) Intravascular delivery of nucleic acid
EP1337242B1 (en) Gene delivery formulations for treatment of ischemic conditions
US7507722B1 (en) Intravascular delivery of nucleic acid
US20050129660A1 (en) Process of delivering a virally encapsulated polynucleotide or viral vector to a parenchymal cell via the vascular system
US20050153451A1 (en) Intravascular delivery of non-viral nucleic acid
US20010008882A1 (en) Process of delivering a polynucleotide into a parenchymal cell

Legal Events

Date Code Title Description
AS Assignment

Owner name: MIRUS BIO CORPORATION, WISCONSIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WOLFF, JON A;BUDKER, TATYANA;REEL/FRAME:021441/0111;SIGNING DATES FROM 20070425 TO 20080815

AS Assignment

Owner name: ROCHE MADISON INC., WISCONSIN

Free format text: CHANGE OF NAME;ASSIGNOR:MIRUS BIO CORPORATION;REEL/FRAME:023081/0128

Effective date: 20080930

Owner name: ROCHE MADISON INC.,WISCONSIN

Free format text: CHANGE OF NAME;ASSIGNOR:MIRUS BIO CORPORATION;REEL/FRAME:023081/0128

Effective date: 20080930

STCF Information on status: patent grant

Free format text: PATENTED CASE

FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FPAY Fee payment

Year of fee payment: 4

AS Assignment

Owner name: ARROWHEAD MADISON INC., WISCONSIN

Free format text: CHANGE OF NAME;ASSIGNOR:ROCHE MADISON INC.;REEL/FRAME:032302/0843

Effective date: 20111114

FPAY Fee payment

Year of fee payment: 8

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20220105